cocaine exposure throughout adolescence

351

Upload: khangminh22

Post on 30-Apr-2023

0 views

Category:

Documents


0 download

TRANSCRIPT

Cecília Juliana Moreira Alves

Cocaine exposure throughout adolescence: a hormonal neurochemical and behavioural study in the rat

Dissertação de Candidatura ao grau de Doutor em Ciências Biomédicas submetida ao Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto Orientador- Professor Doutor Pedro Manuel Ribeiro da Rocha Monteiro Professor Adjunto Escola Superior de Tecnologias da Saúde do Porto, Instituto Politécnico do Porto Co-orientadora- Professora Doutora Liliana Maria de Carvalho e Sousa Professora Associada Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto

Preceitos Legais

Nesta dissertação foram utilizados os resultados dos trabalhos publicados ou em

preparação abaixo indicados. A autora desta dissertação declara que interveio na

concepção e execução do trabalho experimental, na interpretação dos resultados e na

redacção dos resultados publicados ou em preparação, sob o nome de Alves CJ:

In this dissertation were used experimental results published or under preparation stated

below. The author of this dissertation declares that participated in the planification and

execution of the experimental work, in the data interpretation and in the preparation in the

work stated below, under the name Alves CJ:

Publicações em resumo:

Alves CJ, Magalhães A, Summavielle T, de Sousa T, Tavares MA, Monteiro PRR. 2007.

Dopaminergic system response to na anxiogenic situation in rats chronically exposed to

cocaine during adolescence. Neurotoxicology and Teratology 29:402-403.

Alves CJ,

Magalhães A, Summavielle T, Tavares MA, Monteiro PRR. 2006. Effects of

chronic cocaine administration on behavior of adolescent rats in the elevated plus-maze

test. Neurotoxicology and Teratology 28:412- 427.

Publicações por extenso:

Alves CJ, Magalhães A, Summavielle T, Melo P, de Sousa L, Tavares MA, Monteiro

PRR. 2008. Hormonal, neurochemical, and behavioral response to a Forced Swim Test in

adolescent rats throughout cocaine withdrawal. Ann.N.Y.Acad.Sci 1139:366-373.

Alves CJ, Magalhães A, Melo P, de Sousa L, Tavares MA, Monteiro PRR, Summavielle

T. 2009. Cocaine impaired risk-assessment in adolescent rat. (in preparation).

Alves CJ

, Magalhães A, Melo P, de Sousa L, Tavares MA, Summavielle T, Monteiro

PRR. 2009. Altered social behaviour in young adult rats exposed to cocaine throughout

adolescence. (in preparation).

III

Aos meus pais

Agradecimentos

Gostaria de agradecer a todos aqueles que de alguma forma contribuíram para

que a realização deste trabalho fosse possível.

Ao Professor Doutor Pedro Monteiro, meu orientador, que proporcionou a minha

entrada no mundo da ciência, por todo o seu apoio, pela confiança que depositou em

mim e pelo seu optimismo.

À Professora Doutora Liliana de Sousa, minha co-orientadora, por me ter recebido

no seu grupo, pelo apoio e disponibilidade que sempre manifestou para me ajudar, em

especial agradeço a sua ajuda nos estudos comportamentais que integram esta

dissertação, e a sua ajuda com a análise estatística dos resultados.

À Professora Doutora Maria Amélia Tavares, por me ter recebido no seu grupo,

pelo apoio e interesse demonstrados.

À Teresa, por toda a sua ajuda, por encontrar sempre solução para os problemas

que foram surgindo durante a realização deste meu trabalho, por tudo o que me ensinou

e pela sua preocupação com o meu futuro, ficarei para sempre grata.

Ao Pedro e à Ana gostaria de agradecer, mais do que pela sua imensurável ajuda

com trabalho, por tornarem fácil, mesmo em momentos de desânimo, a ida para o

laboratório. À Ana por acreditar em mim, pela sua incansável paciência para me ouvir, e

por tantas vezes me chamar à razão. Ao Pedro pela sua boa disposição, pelos elogios

que fizeram aumentar o meu ego e por me empurrar sempre para a frente. Foi em vós

que tantas vezes fui buscar alento para seguir adiante. Aos dois pelo o que me ajudaram

a crescer durante este período. Para sempre nutrirei uma profunda amizade por vós.

À Joana, por toda a ajuda, em especial por me ter ensinado as técnicas de

dissecção do cérebro indispensáveis à realização deste trabalho.

À Lorena pela sua preocupação com os meus problemas e pelo seu empenho em

tentar ajudar-me sempre, pelos bons momentos e pela amizade.

À Raquel e à Ema que tão importantes foram nos primeiros tempos deste trajecto,

por toda a ajuda.

À Manuela, embora curto o período que passou no laboratório a amizade nasceu

forte e ficará para sempre.

À Aline, à Sílvia, à Danira ao Venâncio e a todos os outros que ao longo destes

anos foram passando pelo laboratório pela vossa ajuda, pela companhia e pela amizade.

VII

Ao Luís e à Nita por todos os fins-de-semana que passaram comigo no IBMC, ao

Luís pela ajuda nas recolhas, à Nita pela ajuda na injecção dos animais, e aos dois pela

vossa amizade.

À minha família, por tudo, por acreditarem em mim, por estarem sempre lá. Vós

sabeis, sois tudo para mim.

À Fundação para a Ciência e a Tecnologia e ao Programa de Financiamento

Plurianual do IBMC pelo apoio financeiro para a execução deste trabalho.

VIII

Abbreviations List

5-HIAA 5-hydroxyindoleacetic acid

5-HT Serotonin ; 5-hydroxytryptamine

5-HT1A Serotonin-1A receptors

5-HT1B Serotonin-1B receptors

5-HT2 Serotonin-2 receptors

5-HT3 Serotonin-3 receptors

AChE Acetylcholinesterase

ACTH Adrenocorticotropic hormone

ANOVA Analysis of variance

AR Androgen receptor

AVP Arginine vasopressin

cAMP cyclic-AMP

CNS Central nervous system

CPP Conditioned place preference

CRH Corticotrophin-releasing hormone

CRH-R1 Corticotrophin-releasing hormone receptor 1

Ct Threshold cycle

CTA Conditioned aversive properties

D1 Dopamine receptor 1

D2 Dopamine receptor 2

D3 Dopamine receptor 3

DA Dopamine

DAT Dopamine transporter

DHT Dihydrotestosterone

DNA Deoxyribonucleic acid

DOPAC 3,4-Dihydroxyphenylacetic acid

e.g. As an example

EPM Elevated plus maze test

IX

Fab Fragment, antigen binding

Fc Fragment, cristallizable

FSH Follicle-stimulating hormone

FST Forced swim test

GABA Gamma-aminobutyric acid

GAPDH Glyceraldahyde-3-phosphate-dehydrogenase

GluR1 Glutamate AMPA receptor subunit GluR1

GnRH Gonadotropin-releasing hormone

GnRH-R Gonadotropin-releasing hormone receptor

GR Glucocorticoid receptor

HPA Hypothalamus-pituitary-adrenals

HPG Hypothalamus-pituitary-gonads

HPLC-EC High-performance liquid chromatography with electrochemical detection

HPT Hypothalamus-pituitary-testes axis

HVA 4-Hydroxy-3-methoxyphenylacetic acid ; homovanillic acid

i.p. Intraperitoneal injection

i.v. Intravenous injection

LH Luteinizing hormone

LHβ Luteinizing hormone β-subunit

LTP Long-term potentiation

MAO Monoamine oxidase

MR Mineralocorticoid receptor

mRNA Messenger ribonucleic acid

mPFC Medial prefrontal cortex

Nac Nucleus accumbens

NR1 Glutamate NMDA receptor subunit NR1

PCR Polymerase chain reaction

PND Postnatal day

POMC Proopiomelanocortin

X

PTSD Post-traumatic stress disorders

PVN Paraventricular nuclei

qRT-PCR Quantitative real-time PCR

SA-HRP Streptavidin-horseradish peroxidase

s.c. Subcutaneous injection

S.E.M. Standard error of the mean

SERT Serotonin transporter

SN Substantia nigra

SN-VTA Ventral mesencephalon (substantia nigra and ventral tegmental area)

TH Tyrosine hydroxilase

UV Ultraviolet

VTA Ventral tegmental area

XI

Resumo

A adolescência é um período em que os sistemas neurobiológicos sofrem

importante reorganização através de processos de produção e eliminação de sinapses,

do desenvolvimento dos sistemas de neurotransmissores e de processos de

mielinização. Durante este período do desenvolvimento, os níveis hormonais alteram-se

de forma drástica em resultado do início da puberdade. Estas alterações reflectem-se

também nas competências cognitivas, psicológicas e sociais. Diversos factores

ambientais podem interferir no desenvolvimento neurobiológico e psico-fisiológico dos

adolescentes. Estudos epidemiológicos mostram que é frequentemente nesta fase que se

iniciam as experiências com drogas de abuso. Paradoxalmente, o estudo dos efeitos da

exposição a drogas de abuso, tem sido realizado principalmente em animais adultos, e

apenas pontualmente no adolescente. No entanto, as características ontogenéticas

desde período e o facto da adolescência representar um período propício para o início do

uso de drogas de abuso, afiguram-se importantes argumentos para que se encare a

adolescência como uma idade chave para o estudo dos efeitos destes compostos. Assim,

com este trabalho pretendeu-se contribuir para a caracterização dos efeitos da cocaína

no adolescente. Para atingir este objectivo foram desenvolvidos vários protocolos

experimentais a fim de caracterizar os efeitos neuroquímicos, hormonais e

comportamentais, a curto e a longo prazo, da exposição crónica a cocaína durante a

adolescência.

Neste trabalho foi utilizado um modelo animal de exposição crónica repetida a

cocaína. Ratos Wistar machos e fêmeas foram tratados diariamente, entre o dia pós-natal

(PND) 35 e o 50, com 3 administrações intraperitoneais de uma solução de hidrocloreto

de cocaína numa concentração de 15 mg/kg de peso corporal. Os grupos controlo foram

injectados com uma dose isovolumétrica de soro fisiológico segundo o mesmo

procedimento experimental. Os efeitos da exposição a cocaína na resposta adaptativa a

um estímulo ansiogénico foram avaliados 2, 5 e 10 dias após retirada da cocaína,

recorrendo-se ao elevated plus maze test (EPM). O comportamento depressivo foi

avaliado 3 dias após a última administração de cocaína, recorrendo-se ao forced swim

test (FST). O comportamento agressivo foi avaliado em ratos machos, 10 dias após a

última administração de cocaína, usando o resident-intruder paradigm. Com o objectivo

de determinar alterações nas respostas neuroquímicas e hormonais aos desafios

proporcionados pelos testes comportamentais, após cada um dos testes foram

determinados, por cromatografia de alta resolução com detecção electroquímica (HPLC-

EC), os níveis dos neurotransmissores dopamina (DA), serotonina (5-HT) e dos seus

XIII

principais metabolitos (DOPAC, ácido 3,4-dihidroxifenilacético; HVA, ácido homovanílico;

5-HIAA, ácido 5-hidroxindolacético), em áreas cerebrais de origem do sistemas

dopaminérgico (SN-VTA, substância negra e área tegmental ventral) e serotoninérgico

(núcleo dorsal da rafe) e em áreas de projecção destes directamente envolvidas nos

efeitos da exposição a drogas de abuso (núcleo accumbens (Nac), estriado dorsal, córtex

pré-frontal, amígdala, hipocampo, hipotálamo e cerebelo). Estes doseamentos foram

também efectuados em condições basais, 30 minutos, 1, 5 e 10 dias após a última

administração de cocaína. Do mesmo modo, os níveis plasmáticos de corticosterona

foram doseados por ensaio imunoenzimático, depois dos testes comportamentais. Ainda

a nível plasmático, foram doseadas, por ensaio imunoenzimático, em ratos macho e

fêmea, as seguintes hormonas: hormona adrenocorticotrópica (ACTH), testosterona,

progesterona e corticosterona, em amostras de sangue recolhidas 30 minutos, 1, 5 e 10

dias após a última administração de cocaína. Os níveis plasmáticos de estradiol foram

também determinados em ratos fêmea nos mesmos pontos de avaliação. Os níveis de

expressão de mRNA para a hormona libertadora da corticotropina (CRH) e receptor de

androgénios (AR) foram medidos no hipotálamo; e os níveis de expressão de mRNA

para: receptor da hormona libertadora de gonadotropinas, receptor da CRH, AR, sub-

unidade β da hormona luteinizante, receptor de glucocorticoides (GR) e

proopiomelanocortina foram determinados na hipófise de ratos machos, 10 dias após o

termo da administração de cocaína, recorrendo à técnica de reacção em cadeia da

polimerase em tempo real (qRT-PCR). Para completar a avaliação de alterações na

actividade do sistema dopaminérgico foram avaliados os níveis de expressão de mRNA

para: tirosina hidroxilase (TH), monoamina oxidase-A, transportador de DA, receptores

dopaminérgicos D1, D2, D3 e de GR em vária regiões cerebrais (SN-VTA, Nac, estriado

dorsal, córtex pré-frontal, amígdala, hipocampo e hipotálamo) 10 dias após a última

administração de cocaína, recorrendo à técnica de qRT-PCR.

Os dados obtidos no estudo comportamental indicam que a exposição a cocaína

durante a adolescência afectou a capacidade de resposta adaptativa a desafios

ambientais. A análise dos resultados mostra que, em ratos macho expostos a cocaína, se

verificam diferenças na capacidade de avaliação de risco durante a execução do EPM,

que são acompanhadas por alterações na neurotransmissão dopaminérgica e

serotoninérgica no córtex pré-frontal e amígdala. Estes animais, apresentam também

sensibilização da resposta do eixo HPA ao estímulo ansiogénico proporcionado pelo

EPM. Estas alterações poderão estar implicadas na diminuição verificada na capacidade

de avaliação de risco perante o estímulo aplicado.

XIV

Na avaliação do comportamento depressivo observado no FST, não se

verificaram diferenças entre grupos. Contudo, os dados neuroquímicos indicam que a

resposta à situação stressante proporcionada pelo FST foi afectada.

Na avaliação dos dados obtidos durante o conflito social proporcionado pelo

resident-intruder paradigm verificou-se que a exposição a cocaína intensificou o

comportamento de investigação social e reduziu o comportamento defensivo, indicando

que a cocaína afectou, em ratos macho, a capacidade de interagir adequadamente com

os conspecificos, no entanto não foram observados efeitos do tratamento no

comportamento agressivo. Estas alterações comportamentais foram suportados por

alterações da actividade dopaminérgica e/ou serotonérgica em áreas cerebrais como a

amígdala, o hipocampo e o córtex pré-frontal.

A análise dos dados hormonais obtidos sugere uma adaptação à exposição

crónica a cocaína, não sendo observadas diferenças nos níveis plasmáticos de ACTH e

de corticosterona 1, 5 e 10 dias após a última administração. No entanto, a activação do

eixo hipotálamo-hipófise-supra-renais (HPA) é evidenciada, em ratos macho, 30 minutos

após a última administração de cocaína, resultando em níveis aumentados de ACTH.

Os dados hormonais obtidos indicam ainda alterações, a curto e a longo prazo, no

funcionamento do eixo hipotálamo-hipófise-gonadas (HPG), também em ratos macho,

verificando-se níveis diminuídos de testosterona 30 minutos e 10 dias após a última

administração de cocaína. Em concordância, 10 dias após a última administração de

cocaína, estes animais apresentavam diminuição do peso relativo das gónadas. Nos

ratos fêmea não foram observadas alterações nos níveis de estradiol ou progesterona,

evidenciado, mais uma vez, uma resposta diferenciada entre géneros.

O estudo neuroquímico em ratos macho, aponta para o desenvolvimento de

tolerância nas respostas dos sistemas dopaminérgico e serotoninérgico aos efeitos

agudos da cocaína após o tratamento crónico durante a adolescência, uma vez que para

a maioria das áreas cerebrais analisadas não foram observadas alterações nos níveis de

DA, 5-HT e seus metabolitos, 30 minutos após a última administração de cocaína.

Contudo, 1 dia após a última administração, verifica-se um aumento da actividade

dopaminérgica no SN-VTA e estriado dorsal, em ratos macho, e na amígdala nas fêmeas.

É proposto que este aumento da actividade dopaminérgica seja resultado do estímulo

produzido pela apresentação de pistas previamente associados à administração de

cocaína, potenciando uma resposta do sistema de recompensa desencadeada pela

expectativa da proximidade da administração. A longo prazo, os dados obtidos no estudo

neuroquímico, revelam um aumento de actividade dopaminérgica na via nigrostriatal, este

XV

aumento é suportado não só pelo aumento dos níveis de DOPAC na SN-VTA e dos

níveis de HVA no estriado dorsal, mas também pelo aumento dos níveis de expressão do

mRNA para a TH na SN-VTA. Nesta região cerebral foi ainda evidenciado aumento dos

níveis de 5-HT. A análise neuroquímica também evidência a resposta diferenciada entre

géneros, verificando-se nas fêmeas, 10 dias após a última administração de cocaína,

aumento de actividade dopaminérgica na amigdala e hipocampo e de actividade

serotoninérgica no hipocampo e Nac.

Os dados apresentados neste estudo mostram que a exposição a cocaína durante

a adolescência induz alterações hormonais, neuroquímicas e comportamentais, a curto e

longo prazo. A comparação dos resultados obtidos neste estudo, com dados da

bibliografia sobre os efeitos da cocaína no adulto, suporta a existência de uma resposta

diferenciada a esta droga durante a adolescência. Salientam-se, na presente dissertação,

as diferenças observadas entre ratos adolescentes macho e fêmea, após exposição

crónica a cocaína, a nível comportamental, neuroquímico e hormonal. Simultaneamente,

este trabalho mostra, pela primeira vez, que a exposição crónica a cocaína na

adolescência compromete, em ratos macho, a adequação da resposta comportamental a

um estímulo ansiogénico, o que é suportado pelos parâmetros fisiológicos avaliados. Os

dados apresentados contribuem de forma relevante para a clarificação dos factores

envolvidos na maior susceptibilidade à experimentação de drogas de abuso evidenciada

na adolescência.

XVI

Abstract

Throughout adolescence, the neurobiological systems are still undergoing

important development rearrangements, associated to continuous plastic events that

result from an integrated process of overproduction and elimination of synapses, evolution

of the neurotransmitter systems and progressive myelination. Hormonal levels are also

changed dramatically during this period as a result of the onset of puberty. Concomitant to

these changes in the brain and hormonal status, significant transitions occur in cognitive,

psychological and social competences. At this time, environmental factors may have a

strong impact on the unique neurobiological and psycho-physiological profile of

adolescents. Epidemiological data indicate that experiences with drugs of abuse and the

onset of addictive disorders are primarily concentrated in the adolescence and early

adulthood. However, studies on the effects of drugs of abuse, have primarily been

conducted in adult animal models and little is known regarding the effects that these drugs

on the adolescent. The objective of the present study was to contribute to further

characterize the effects of exposure to drugs of abuse at this key developing time, using

an animal model of exposure to cocaine. Experimental protocols were developed to

assess short and long-term neurochemical, hormonal and behavioural effects of chronic

cocaine exposure, throughout adolescence, in a rat model.

Male and female Wistar rats were daily injected intraperitoneally with 3 doses of 15

mg/kg per body weight of cocaine hydrochloride, in 0.9% NaCl solution, from postnatal

day (PND) 35 to PND 50. Controls were given isovolumetric 0.9% NaCl solution following

the same experimental protocol. The anxiety-like behaviour was evaluated using the

elevated plus maze test (EPM) in male rats, 2, 5 and 10 days after cocaine withdrawal,

and in female rats, 2 days after withdrawal. The depressive-like behaviour was evaluated

using the forced swim test (FST) both in male and female rats, 3 days after cocaine

withdrawal. The aggressive behaviour was evaluated in male rats, 10 days after

withdrawal, using the resident-intruder paradigm. The levels of neurotransmitters were

analysed after each behavioural test and at 30 minutes, 1, 5 and 10 days after the last

administration of cocaine, through measuring levels of dopamine (DA), serotonin (5-HT)

and its metabolites (3,4-dihydroxyphenylacetic acid, DOPAC; homovanilic acid, HVA; 5-

hydroxyindoleacetic acid, 5-HIAA), by high performance liquid chromatography with

electrochemical detection (HPLC-EC), in the ventral mesencephalon (SN-VTA), the dorsal

raphe nucleus, the nucleus accumbens (Nac), the dorsal striatum, the prefrontal cortex,

the amygdala, the hippocampus, the hypothalamus and the cerebellum. The plasma

levels of adrenocorticotropic hormone (ACTH), corticosterone, testosterone and

XVII

progesterone were determined by an enzyme immunoassay, both in male and female rats

at 30 minutes, 1, 5 and 10 days after the last administration of cocaine. In females, the

levels of estradiol were also determined. Levels of corticosterone were assessed, as well,

after each behavioural challenge. Moreover, in male rats, the expression levels of mRNA

transcripts for corticotrophin-releasing hormone (CRH) and the androgen receptor (AR)

were measured in the hypothalamus; while the expression levels of mRNA transcripts for

CRH receptor 1, gonadotropin-releasing hormone receptor, AR, luteinizing hormone β-

subunit, glucocorticoid receptor (GR) and proopiomelanocortin were measured in the

pituitary, 10 days after cocaine withdrawal, by quantitative real-time polymerase chain

reaction (qRT-PCR). The expression levels of mRNA transcripts, 10 days after cocaine

withdrawal, for tyrosine hydroxylase (TH) were measured by qRT-PCR in the SN-VTA,

Nac and dorsal striatum, while the expression levels of mRNA transcripts for monoamine

oxidase-A, DA transporter, DA receptors (D1, D2, D3) and GR were measured in the SN-

VTA, Nac, dorsal striatum, prefrontal cortex, amygdala, hippocampus and hypothalamus.

The most relevant results obtained indicate that chronic cocaine exposure through

adolescence negatively interfered with the adaptive responses to environmental

challenges. Data analysis showed impaired risk-assessment during the EPM apparatus

exploration, both 2 and 10 days after withdrawal, which was supported by impaired

dopaminergic and serotonergic neurotransmission in the prefrontal cortex and impaired

dopaminergic neurotransmission in the amygdala. These neurochemical alterations may

be involved in the failure of cocaine-treated male rats to exhibit an adaptive behaviour

during the EPM. Cocaine treatment did not have a significant effect on depressive-like

behaviour in both male and female rats, as assessed 3 days after withdrawal. However,

when exposed to the stressful situation provided by the FST, an altered response was

observed in the neurochemical data. Moreover, although no long-term effects of cocaine

were observed in the aggressive behaviour of male rats, as assessed 10 days after

withdrawal, cocaine-exposed males intensified the social investigation behaviour and

reduced the defensive behaviour during the social conflict provided by the resident-

intruder paradigm, indicating that cocaine affected the ability of young adult rats to

properly interact with conspecifics. In accordance, the dopaminergic and the serotonergic

activities, in response to this social conflict, were altered in cocaine-exposed animals.

The neurochemical data also showed that cocaine exposure throughout

adolescence produced long-lasting changes upon the monoamine systems. In male rats,

this was reflected by increased dopaminergic activity in the nigrostriatal pathway,

supported by the increased levels of DOPAC in SN-VTA, increased levels of HVA in

dorsal striatum and increased expression levels of TH mRNA in the SN-VTA; and long-

XVIII

lasting increased 5-HT levels in SN-VTA. In cocaine-treated females, these alterations

were revealed through increased dopaminergic activity in amygdala and in hippocampus,

and increased serotonergic activity in hippocampus and Nac.

The analysis of the hormonal data indicates an activation of the hypothalamus-

pituitary-adrenal (HPA) axis in male rats, 30 minutes after the last administration of

cocaine, as showed by the increased ACTH levels. Despite this acute effect after chronic

cocaine treatment, the basal activity of the HPA axis did not appear to have been affected,

as no changes were observed in ACTH and corticosterone levels at 1, 5 and 10 days after

withdrawal, in both male and female rats. Nevertheless, a disruption of the HPA axis

response under anxiogenic conditions was revealed in young adult male rats, which

displayed a sensitization of the corticosterone response to the EPM when tested 10 days

after withdrawal. Importantly, this response was accompanied by an apparent loss of risk

assessment abilities.

Globally, data presented in this study showed that chronic cocaine exposure

throughout adolescence produced hormonal, neurochemical and behavioural short and

long-term alterations. The present study highlights important gender difference in the

behavioural, neurochemical and hormonal effects of chronic cocaine exposure throughout

adolescence. Simultaneously, it was shown that adolescent male rats exposed to cocaine,

fail to exhibit an adaptive behavioural response to an anxiogenic stimulus, which was

paralleled by alterations in physiologic parameters. Moreover, compared to published data

our results seem to indicate that adolescent rats respond to chronic cocaine treatment

differently than adult animals, and extensively contributes to the growing understanding of

the mechanisms involved in the increased susceptibility to drug abuse during

adolescence.

XIX

Résumé

Pendant l'adolescence, les systèmes neurobiologiques continuent à présenter des

importants réarrangements, associés à des événements plastiques résultants d'un

processus intégré de surproduction et l'élimination des synapses, évolution des systèmes

de neurotransmetteurs et progressive myélinisation. Les niveaux hormonaux change

également au cours de cette période. Concomitante de ces changements dans le cerveau

et le statut hormonal, d'importantes transitions arrivent dans les compétences cognitives,

psychologiques et sociales. À ce moment, les facteurs environnementaux peuvent avoir

un impact fort sur le profil neurobiologique et psycho-physiologique unique des

adolescents. Les données épidémiologiques indiquent que les expériences avec les

drogues et l'apparition des troubles de dépendance sont essentiellement concentrées

dans l'adolescence et au début de l'âge adulte. Toutefois, des études sur les effets de

l'abus des drogues, ont, principalement, été effectuées parmi des modèles animaux en

adultes. L'objectif de cette étude était de contribuer à caractériser les effets de l'exposition

aux drogues à ce moment du développement, en utilisant un modèle animal de

l'exposition à la cocaïne. Des protocoles expérimentaux ont été développés afin d'évaluer

à court et à long terme les effets neurochimiques, hormonaux et comportamentaux de

l'exposition chronique à la cocaïne, au cours de l'adolescence dans un modèle animal un

rat.

Des mâles et des femelles des rats Wistar ont été administrées, par jour et par

voie intrapéritonéale, 3 doses de 15 mg/kg de poids corporel de chlorhydrate de cocaïne,

dans une solution 0,9% NaCl, du jour postnatal (PND) 35 au PND 50. Les contrôles ont

reçu une solution isovolumetric NaCl 0,9% suivant le même protocole expérimental. Le

comportement d’anxiété a été évalué en utilisant le elevated plus maze test (EPM) chez

les rats mâles, 2, 5 et 10 jours après le dernier apporte de cocaïne, et chez les rats

femelles, 2 jours après. Le comportement dépressif a été évalué à l'aide du forced swim

test (FST) dans les rats mâles et femelles, 3 jours après le retrait de cocaïne. Le

comportement d’agression a été évalué chez les rats mâles, 10 jours après le retrait, en

utilisant le resident-intruder paradigm. Les niveaux de neurotransmetteurs ont été

analysés après chaque test de comportement et à 30 minutes, 1, 5 et 10 jours après la

dernière administration de cocaïne, par la mesure des niveaux de dopamine (DA),

sérotonine (5-HT) et de ses métabolites (acide 3,4-dihydroxyphenylacétique, DOPAC;

acide homovanilique, HVA; acide 5-hydroxyindole acétique, 5-HIAA), par

chromatographie liquide d’haute performance avec détection électrochimique (HPLC-EC),

dans le mesencephale ventral (VTA-SN), le noyau raphé dorsal, le nucleus accumbens

XXI

(Nac), le striatum dorsal, le cortex préfrontal, l'amygdale, l'hippocampe, l'hypothalamus et

le cervelet. Les niveaus plasmatiques de l'hormone adrénocorticotrope (ACTH), de la

corticostérone, de la testostérone et de la progestérone ont été déterminés dans les rats

mâles et femelles à 30 minutes, 1, 5 et 10 jours après la dernière administration de

cocaïne. Chez les femelles, les niveaux de l'estradiol ont également été déterminés. Les

niveaux de corticostérone ont été aussi évalués après chaque défi de comportement. En

outre, chez les rats mâles, les niveaux d'expression de l'mRNA des transcriptions pour l’

hormone de libération de la corticotrophine (CRH) et le récepteur des androgènes (AR)

ont été mesurés dans l'hypothalamus, alors que le niveau d'expression de l'mRNA des

transcriptions pour récepteur CRH-1, récepteur de la hormone de libération des

gonadotrophines, AR, hormone lutéinisante sous-unité β, ré cepteur des glucocorticoïdes

(GR) et proopiomélanocortine ont été mesurés dans l'hypophyse, 10 jours après le retrait

de cocaïne, par la réaction quantitative en chaîne de la polymérase en temps réel (qRT-

PCR). Les niveaux d'expression des transcriptions de l'mRNA, 10 jours après le retrait de

cocaïne, de la tyrosine hydroxylase (TH) ont été mesurés par qRT-PCR dans le SN-VTA,

Nac et striatum dorsal, tandis que les niveaux d'expression de l'mRNA transcrits pour la

monoamine-oxydase A, DA transporteur, DA récepteurs (D1, D2, D3) et des GR ont été

mesurés dans le SN-VTA, Nac, striatum dorsal, cortex préfrontal, l'amygdale,

l'hippocampe et l'hypothalamus.

Les résultats les plus pertinents obtenus indiquent que l'exposition chronique de

cocaïne pendant l'adolescence perturbe négativement les réponses adaptatives aux défis

environnementaux. L'analyse des données a montré facultés d'évaluation des risques lors

de l' apparat d'exploration de l’ EPM, 2 et 10 jours après le retrait, qui a été appuyée par

une altération de la neurotransmission dopaminergique et sérotoninergique dans le cortex

préfrontal et l'altération de la neurotransmission dopaminergique dans l'amygdale. Ces

modifications neurochimiques peuvent être impliqués dans la faillite des rats mâles en

afficher un comportement adaptatif au cours de l'EPM. Le traitement avec cocaïne n'a pas

eu d'effet significatif sur le comportement dépressif dans les rats mâles et femelles,

d’après évaluation 3 jours après le retrait. Toutefois, lorsqu'il est exposé à la situation de

stress par la FST, une altération de la réponse a été observée dans les données

neurochimiques. En outre, même si aucun effet à long terme de la cocaïne a été observé

dans le comportement agressif des rats mâles, d’après évaluation 10 jours après le retrait,

les mâles exposés à cocaïne ont intensifié le comportement de recherche sociale et

diminué défensive comportement au cours des conflits sociaux prévues par le resident-

intruder paradigm, ce qui indique que cocaïne a affecté la capacité des jeunes rats

adultes de bien interagir avec leurs congénères. Conformément, les activités

XXII

dopaminergiques et sérotoninergiques, en réponse à ce conflit social, ont été modifiées

dans animaux exposés à cocaïne.

Les données neurochimiques de cocaïne ont montré également que l'exposition à

la cocaïne pendant l'adolescence produit des changements durables aux systèmes

monoamine. Chez les rats mâles, ça s'est traduit par une augmentation de l'activité

dopaminergique dans la voie nigrostriatal, soutenue par l'augmentation des niveaux de

DOPAC au SN-VTA, l’augmentation des niveaux d'HVA dans le striatum dorsal et

l'augmentation des niveaux d'expression des mRNA de la TH dans le SN-VTA et niveaux

durables et augmentés de 5-HT dans le SN-VTA. Dans les femelles traitées à la cocaïne,

ces modifications ont été révélés grâce à une augmentation de l'activité dopaminergique

dans l'amygdale et l'hippocampe, et l'augmentation de l'activité sérotoninergique dans

l'hippocampe et Nac.

L'analyse des données hormonales indique une activation de l'axe de

l'hypothalamus-hypophyso-surrénalien (HPA) dans les rats mâles, 30 minutes après la

dernière administration de la cocaïne, comme l'a montré l'augmentation des niveaux

d'ACTH. Malgré ce grave effet chez les rats mâles, l'activité basale de l'axe HPA ne

semble pas avoir été touchés par le traitement avec de la cocaïne, comme aucun

changement n'a été observé dans l'ACTH et dans les niveaux de corticostérone 1, 5 et 10

jours après le retrait, chez les rats mâles et femelles. Néanmoins, une rupture de la

réponse de l'axe HPA sous des conditions anxiogéniques a été révélé chez les jeunes-

adultes rats mâles, en faisant preuve d'une sensibilisation de la réponse à la

corticostérone EPM lors d'un essai 10 jours après le retrait. Surtout, cette réponse a été

accompagnée par une apparente perte de capacités d'évaluation des risques, qui est

renforcée par la baisse des niveaux de testostérone observée aussi bien à 30 minutes et

10 jours après la dernière administration de cocaïne.

Généralement, les données présentées dans cette étude ont montré que

l'exposition chronique à la cocaïne pendant l'adolescence produit des modifications

hormonales, neurochimiques et comportamentales à court et à long terme. La présente

étude met en évidence une importante différence entre les sexes en ce qui concerne les

effets comportamentaux, hormonaux et neurochimiques de l'exposition chronique à la

cocaïne, au cours de l'adolescence. Simultanément, il a été démontré que les rats mâles

adolescents exposés à la cocaïne, n’ont pas été en mesure de présenter une réponse

comportamentale adaptative à un stimulus anxiogène, qui se traduit par des modifications

de paramètres physiologiques. De plus, nos résultats par rapport aux données publiées,

semblent indiquer que les rats adolescents répondent différemment des animaux adultes

à un traitement chronique de cocaïne. Ce travail contribue largement à la compréhension

XXIII

des mécanismes impliqués dans la susceptibilité des adolescents à l’égard de

l'expérience de la drogue.

XXIV

Este trabalho foi co-financiado pela Fundação para a Ciência e Tecnologia através de

uma bolsa de doutoramento (SFRH/BD/17195/2004) e pelo Programa de Financiamento

Plurianual do IBMC.

This work was funded by Fundação para a Ciência e Tecnologia through a PhD fellowship

(SFRH/BD/17195/2004) and by the Programa de Financiamento Plurianual do IBMC.

XXV

Contents

Chapter 1- Introduction

1.1. General 3

1.2. Neuropharmacology of cocaine 4

1.3. Addiction: the transition to compulsive drug seeking 6

1.4. Cocaine and the Dopaminergic and Serotonergic Systems 8

1.4.1. Effects of cocaine upon the dopaminergic system 8

1.4.2. Effects of cocaine upon the serotonergic system 10

1.5. Cocaine and the Neuroendocrine System 12

1.5.1. The Hypothalamus-Pituitary-Adrenal axis 12

1.5.1.1. Effects of cocaine upon the HPA axis activity 14

1.5.1.2. The HPA axis and the behavioural effects of cocaine 17

1.5.1.2.1. Stress and increased vulnerability to cocaine 18

1.5.1.2.1.1. Interaction between stress, glucocorticoids and vulnerability to drugs 20

1.5.2. The Hypothalamus-Pituitary-Gonadal axis 21

1.5.2.1. The Hypothalamus-Pituitary-Testicular axis 21

1.5.2.2. The Hypothalamus-Pituitary-Ovaries axis 23

1.5.2.2.1. Estrous cycle 25

1.5.2.3 Effects of cocaine on the HPG axis 26

1.5.2.4. Effects of sex and sex hormones in the response to cocaine 28

1.5.3. Interactions HPG/ HPA axes and the response to cocaine 31

1.6. The adolescence 34

1.6.1. Adolescence: A critical period of addiction vulnerability 37

1.7. Objectives 43

1.8. References

45

Chapter 2- Methods

2.1. Animal model 69

2.1.1. Introduction 69

XXVII

2.1.2. Animals 71

2.1.3. Chronic administration and assessment time points 72

2.2. Blood and tissue collection 73

2.3. Hormone plasma levels measures 73

2.3.1. Steroid plasma levels 73

2.3.2. ACTH plasma levels 74

2.4. Neurotransmitters determinations 75

2.5. mRNA expression levels determination 76

2.6. Behavioural tests 77

2.6.1. Elevated plus maze test 77

2.6.2. Forced swim test 77

2.6.3. Aggressiveness test 78

2.7. Statistical analysis 79

2.8. References

80

Chapter 3- Short-term effects of cocaine exposure throughout adolescence in rat

3.1. Introduction 85

3.2. Material and Methods 87

3.2.1. Animals 87

3.2.2. Behavioural tests 87

3.2.2.1. Elevated plus maze test 87

3.2.2.2. Forced swim test 87

3.2.3. Blood and tissue collection 88

3.2.4. Hormone plasma levels determinations 88

3.2.5. Neurochemical determinations 89

3.2.6. Statistical analysis 89

3.3. Results 90

3.3.1. Analyses performed 30 minutes and 1 day after the last administration of cocaine 90

XXVIII

3.3.1.1. Biometric measures 90

- Evolution of body weight 90

- Adrenal weight 90

- Gonadal weight 91

3.3.1.2. Hormone plasma levels 92

- ACTH 92

- Corticosterone 92

- Testosterone 92

- Progesterone 92

- Estradiol 92

3.3.1.3. Neurochemical determinations 95

3.3.2. Behavioural studies and associated measures 111

3.3.2.1. Anxiety-like behaviour 111

- Elevated plus maze test 111

- Corticosterone plasma levels 114

- Neurochemical determinations 115

3.3.2.2. Depressive-like behaviour 125

- Forced swim test 125

- Corticosterone plasma levels 126

- Neurochemical determinations 126

3.4. Discussion 137

3.4.1. Effects of chronic cocaine treatment during adolescence on the body weight evolution 137

3.4.2. Effects of chronic cocaine treatment during adolescence on the neuroendocrine function 137

3.4.3. Effects of chronic cocaine treatment during adolescence on the dopaminergic and serotonergic systems function 144

3.4.4. Effects of chronic cocaine treatment during adolescence on the anxiogenic-like behaviour 159

3.4.5. Effects of chronic cocaine treatment during adolescence on the depressive-like behaviour 164

3.5. Conclusion 166

XXIX

3.6. References

168

Chapter 4- Long-term effects of cocaine throughout adolescence in rat

4.1. Introduction 187

4.2. Material and Methods 189

4.2.1. Animals 189

4.2.2. Behavioural tests 189

4.2.2.1. Elevated plus maze test 189

4.2.2.2. Resident-intruder paradigm 189

4.2.3. Blood and tissue collection 190

4.2.4. Hormone plasma levels determinations 190

4.2.5. Neurochemical determinations 191

4.2.6. mRNA expression levels determination 191

4.2.7. Statistical analysis 193

4.3. Results 193

4.3.1. Basal analyses 195

- Evolution of body weight 195

- Adrenal weight 195

- Gonadal weight 196

4.3.1.2. Hormone plasma levels 197

- ACTH 197

- Corticosterone 197

- Testosterone 197

- Progesterone 197

- Estradiol 198

4.3.1.3. Neurochemical determinations 199

4.3.1.4. mRNA expression levels determination 215

XXX

4.3.1.4.1. Dopaminergic mRNA-related expression levels 215

4.3.1.4.2. HPA and HPG axes mRNA-related expression levels 222

4.3.2. Behavioural studies and associated measures 225

4.3.2.1. Anxiety-like behaviour 225

- Elevated plus maze test 225

- Corticosterone plasma levels 227

- Neurochemical determinations 228

4.3.2.2. Aggressive behaviour 238

- Resident-intruder paradigm 238

- Corticosterone plasma levels 242

- Neurochemical determinations 242

4.4. Discussion 253

4.4.1. Effects of chronic cocaine treatment during adolescence and withdrawal on the body weight evolution 253

4.4.2. Effects of chronic cocaine treatment during adolescence and withdrawal on the neuroendocrine function 253

4.4.3. Effects of chronic cocaine treatment during adolescence on the dopaminergic and serotonergic systems function 258

4.4.4. Effects of chronic cocaine treatment during adolescence on the anxiogenic-like behaviour 271

4.4.5. Effects of chronic cocaine treatment during adolescence on the aggressive behaviour 276

4.5. Conclusion 282

4.6. References

285

Chapter 5- General conclusion

5. General conclusion

303

Appendix I- Published articles 307

Article- Alves CJ

309

, Magalhães A, Summavielle T, Melo P, de Sousa L, Tavares MA, Monteiro PRR. 2008. Hormonal, neurochemical, and behavioral response to a Forced Swim Test in adolescent rats throughout cocaine withdrawal. Ann.N.Y.Acad.Sci 1139:366-373.

XXXI

Chapter 1

Introduction

1.1. General

Cocaine is a psychostimulant drug, obtained from the leaves of the plant

Erythroxylum coca. For thousand years the natives of South America have chewed the

coca leaf to increase strength and energy, but the europeans did not hear about this plant

until the Spanish colonized South America in the XVI century (Karch, 1996). In the early

1560s, Nicholas B. Monardes published a book describing the practice of the natives of

chewing coca leaves to induce “great contentment”, at the time he was already aware that

coca had also undesirable side effects (Karch, 1996).

Isolation of cocaine, the coca’s active principle, was first described by Albert

Niemann in his Ph.D. dissertation titled “On a New Organic Base in the Coca Leaves”,

which was published in 1860 (Karch, 1996). For a long time the medical community

remained unimpressed with coca and even after the purification of cocaine, interest in its

therapeutic applications remained slight. However, in 1884, with the publication of

Freud’s paper “Über Coca” and the discovery of cocaine’s local anesthetic properties by

Köller, the interest of the medical community in this alkaloid increased (Karch, 1996).

Cocaine was propelled into the limelight and physicians around the world were soon

experimenting cocaine in a wide range of conditions (Karch, 1996).

The first reports of cocaine toxicity appeared less than one year after Köller’s and

Freud’s publications, and during the 1880´s several reports were published describing

toxic reactions associated with cocaine and cocaine-related deaths (Karch, 1996).

However, none of these negative reports appeared to have much impact. The popularity

of coca was not affected and thousands of cocaine-containing patented medicines flooded

the market, some with truly enormous amounts of cocaine (Karch, 1996).

During the early years of the last century it became evident that cocaine was

addicting and could produce serious medical complications, especially with the availability

of cocaine powder for intranasal or intravenous use. In 1906 cocaine became illegal in the

United States and its use waned (Karch, 1996). Between 1924 and 1973, there was only

one reported fatality and it involved a surgical misadventure, this absence of case reports

reflected a decline in its use, probably related with the outlawing of cocaine and with the

appearance of amphetamines (Dackis and O'Brien, 2001).

In the 1960s cocaine resurfaced with a mythology of safety and glamour, and this

perception encouraged millions of first-time users, forming the pool from which cocaine-

addicted individuals emerged (Dackis and O'Brien, 2001). With the appearance of “crack”

cocaine in 1986, another order of magnitude increase in dosage occurred and cocaine-

3

related illness became a significant cause of morbidity and mortality (Dackis and O'Brien,

2001).

Presently, according to the 2008 World Drug Report of the Office of Drug and

Crime of the United Nations, about 5% of the population between the ages of 15 and 64

use illicit drugs, which represents about 200 million people (UNODC, 2008). Cocaine is

between the most used drugs, with 16 million of users (UNODC, 2008).

The 2008 World Drug Report of the Office of Drug and Crime of the United Nations

reported that the cocaine consumer market in North America contracted, however the

consumption increased in South America, Western Europe and West and Southern Africa

(UNODC, 2008). The largest numbers of cocaine users are found in North America (7.1

million people), followed by West and Central Europe (3.9 million), and South America

(including Central America and the Caribbean: 3.1 million) (UNODC, 2008).

Most of the global increase of cocaine use over the last decade can be attributed

to rapidly rising cocaine consumption in Europe (UNODC, 2008). Increases in cocaine use

in 2006 were reported by a number of South-European countries, notably Spain, Portugal,

Italy and some countries of the western Balkan, as well as France, the United Kingdom,

Ireland and several Nordic countries (UNODC, 2008). The highest prevalence rates for

cocaine use in Europe are found in Spain, where cocaine use doubled among the general

population (age 15-64), from 1.6% in 1999 to 3.0% in 2005. In fact cocaine use levels in

Spain are similar to those reported from the USA (UNODC, 2008).

A major problem in what concerns to cocaine use is its consumption by young

people. In Europe, population surveys carried out in a number of countries have recorded

a marked increase in use among young people since the mid-1990s (EMCDDA, 2008).

1.2. Neuropharmacology of cocaine

Cocaine possesses potent psychostimulant properties as measured by antifatigue

and stimulant actions (Fischman et al., 1983; Romach et al., 1999). In rodents, cocaine

increases locomotor activity (Bhattacharyya and Pradhan, 1979; Abel et al., 1989) and

decreases food intake (Balopole et al., 1979). Cocaine has also a high abuse potential,

and experimental studies have shown that cocaine induces place preference conditioning

(Spyraki et al., 1982) and readily acts as reinforcer for drug self-administration (Roberts

and Koob, 1982; Goeders et al., 1993).

4

Psychostimulants of high abuse potential, such as cocaine and amphetamine,

interact initially to block monoamine transporter proteins which are located on

monoamonergic nerve terminals (Glowinski and Axelrod, 1965; Ferris et al., 1972;

Iversen, 1973; Ritz et al., 1987). Cocaine inhibits, with about equal potency, the uptake of

the three major monoamines neurotransmitters, dopamine (DA), serotonin (5-HT) and

norepinephrine (NE) (Rothman et al., 2001), thereby, potentiating monoaminergic

transmission (Pettit and Justice, 1989; Broderick et al., 2003).

However, the primary neuropharmacological action responsible for its psychomotor

stimulant and reinforcing effects appears to be on the dopaminergic systems in the central

nervous system (CNS) (Koob, 1992a). Antidepressant drugs that block NE and/or 5-HT,

but not DA reuptake, produce neither significant reinforcement in animal models nor

euphoria in humans (Hyman, 1996). Adrenoceptor antagonists, such as

phenoxybenzamine and phentolamine also have no effects on cocaine reinforcement,

while on the other hand, DA receptor antagonists block cocaine reinforcement (De Wit

and Wise, 1977). In addiction, lesion of DA terminals within the nucleus accumbens (Nac)

or full pharmacological blockade of DA receptors inhibits cocaine self-administration

(Roberts et al., 1980).

Brain dopaminergic neurons are organized into two major pathways that originate

in the midbrain and project to numerous forebrain and cortical regions. The

mesocorticolimbic dopaminergic system, projects from the ventral tegmental area (VTA) to

the ventral forebrain, including the Nac, olfactory tubercle, frontal cortex, amygdala and

septal area. The nigrostriatal dopaminergic system arises primarily from the substantia

nigra (SN) and projects to the corpus striatum. This last dopaminergic pathway is

implicated in the focused repetitive behaviour associated with psychostimulants abuse,

called stereotyped behaviour (Creese and Iversen, 1974; Robbins and Everitt, 1996). The

mesocorticolimbic dopaminergic system has been primarily implicated in cocaine-induced

locomotion and in the reinforcing effects of cocaine (Everitt and Wolf, 2002).

The mesolimbic dopaminergic system and its forebrain targets are very old from an

evolutionary point of view and are part of the motivational system that regulates

responses to natural reinforcers such as food, drink, sex and social interaction (for review

see Nestler, 2001). Drugs of abuse affect the brain reward system with a strength and

persistence not seen in natural reinforcers (Nestler, 2001; Robinson and Berridge, 2003).

In particular, the mesoaccumbens dopaminergic pathway, extending from the VTA

to the Nac, a brain region thought to be involved in converting emotion into motivated

action and movement (for review see Mogenson et al., 1980), seems to be where cocaine

5

act to produce its acute reinforcing actions (Hyman, 1996). Blockage of cocaine induced

locomotor activation has been observed following 6-hydroxydopamine lesions of the

region of the Nac (Le Moal and Simon, 1991; Sellings et al., 2006). Lesions of the Nac

also produce disruption of cocaine self-administration (Roberts et al., 1980; Pettit et al.,

1984; Zito et al., 1985). However, 6-hydroxydopamine lesions of the frontal cortex and

caudate nucleus fail to significantly alter established cocaine self-administration (Martin-

Iverson et al., 1986). Neurochemical studies using in vivo studies demonstrate that

cocaine increases extracellular DA to a greater extent in the Nac (Carboni et al., 1989;

Pettit and Justice, 1989; Cass et al., 1992).

Figure 1.2.1- Sagittal section through a representative rodent brain illustrating the dopaminergic pathways

implicated in psychomotor stimulant and reinforcing actions of cocaine. Nac- nucleus accumbens; SN-

substantia migra; VTA- ventral tegmental area.

1.3. Addiction: the transition to compulsive drug seeking

The drug addiction process is normally accepted to be composed of four major

phases: initiation, during which drug consumption is relatively low and irregular;

maintenance, during which frequent and compulsive intake of large amounts of drugs is

observed; withdrawal, during which the individual attempts to quit drug self-administration;

and relapse, during which the individual resumes compulsive drug use (Sarnyai et al.,

2001).

The acute reinforcing effects of cocaine lead to patterns of drug use that, in

epigenetically vulnerable individuals, eventually result in addiction (Hyman, 1996). Drug

addiction is a chronically relapsing disorder characterized by the compulsion to seek and

take a drug, with loss of control in limiting intake (Nestler, 2001). Addiction is also

6

characterized by the emergence of a negative emotional state (e.g., dysphoria, anxiety,

irritability) when access to the drug is prevented (Koob and Moal, 2006).

Drug-induced neuroadaptations are thought to be critical in the transition to

addiction. Extensive studies both in animal models of a variety of species, as well as basic

clinical research in humans, using neurochemical, molecular and related behavioural

technologies, as well as a variety of imaging techniques, have documented that indeed

chronic exposure to drugs does cause alterations in specific aspects of brain function

which are persistent over varying periods of time or, in some cases, may even be

permanent (Kreek and Koob, 1998; Nestler, 2001).

As cocaine use and duration increases, the positive reinforcing effects are

diminished while the dysphoria (including agitation, anxiety and even panic attacks)

increase, suggesting acute tolerance to the arousing and positive mood effects of cocaine

(Koob and Moal, 2006). Tolerance can be defined as a given drug producing a decreasing

effect with repeated dosing or when larger doses must be administered to produce the

same effect (Koob and Moal, 2006). Tolerance to the reinforcing effects of cocaine may

be marked, leading to administration of very high drug doses (Hyman, 1996). However,

tolerance does not develop to the stereotyped behaviour and psychosis induced by

stimulants and, in fact, these behavioural effects appear to show a sensitization (Post et

al., 1992; Hyman, 1996). Sensitization being defined as the long-lasting increment in

response occurring upon repeated presentation of a stimulus (Nestler, 2001).

The neuroadaptative processes of tolerance and sensitization together with

withdrawal symptoms have been proposed as key elements in the development of

addiction (Hyman and Malenka, 2001). Withdrawal signs associated with cessation of

chronic drug administration usually are characterized by responses that are opposite to

the initial effects of the drugs. Withdrawal from chronic or high dose cocaine use in

humans is associated with relatively few overt physical signs but a number of

motivationally relevant symptoms such as dysphoria and depression, anxiety, anergia,

insomnia and craving for the drug (a compelling desire to re-experience the cocaine

experience (Koob and Caine, 1999)) (Gawin and Kleber, 1986; Weddington et al., 1990;

Satel et al., 1991; Miller et al., 1993).

There are several theoretical explanations of how drug-induced alterations in

psychological function might cause a transition to addiction. The most traditional

theoretical explanation of how drug–induced alterations might cause transition to addiction

is the hedonic view that drug pleasure and subsequent unpleasant withdrawal symptoms

are the chief causes of addiction (Robinson and Berridge, 2003). It defends that drugs are

7

taken first because they are pleasant, but with repeated drug use homeostatic

neuroadaptations lead to tolerance and addiction, such that unpleasant withdrawal

symptoms ensue upon the cessation of use, thus, compulsive drug taking is maintained,

to avoid unpleasant withdrawal symptoms (Koob and Moal, 1997). It is suggest that

repeated drug use induces tolerance or downregulation in the mesolimbic dopaminergic

system, decreasing the pleasant drug “highs”, sudden cessation of drug use causes

dopaminergic (and serotonergic) neurotransmission to further drop below normal levels, at

least for several days, resulting in the dysphoric state of withdrawal (Koob and Moal,

1997). This theoretical explanation also suggest that repeated drug use can activate brain

and hormonal stress responses (Koob and Moal, 2006). As a result, addicts who originally

take drugs to gain a positive hedonic state are spiraled into a predominantly negative

hedonic state, which causes the transition to addiction (Koob and Moal, 1997, 2006).

1.4. Cocaine and the Dopaminergic and Serotonergic Systems

1.4.1 Effects of cocaine upon dopaminergic system

As mentioned above, the increase in extracellular DA following cocaine

administration plays a major role in cocaine reinforcement. Studies using microdialysis

have shown that acute cocaine administration leads to an immediate and dose-dependent

increase in extracellular DA levels (Church et al., 1987; Carboni et al., 1989; Maisonneuve

and Kreek, 1994). Increased synaptic levels of DA will stimulate DA receptors and activate

several signal transduction pathways. Pharmacological studies with selective D1, D2 and

D3 DA receptors antagonists and knockout studies have shown that all three receptors

subtypes appear to mediate the reinforcing effects of cocaine, albeit possibly different

components of the response. Low doses of D1 DA receptor antagonist block the

reinforcing effects of intravenous cocaine self-administration (Maldonado et al., 1993;

Caine and Koob, 1994). D2 DA receptor antagonists block responding for cocaine but also

have pronounced motor response inhibitory actions (Koob and Moal, 2006). D3 DA

receptor antagonists block drug-seeking behaviour associated with cocaine in second-

order and progressive-ratio schedules (Koob and Moal, 2006).

Many studies have shown that chronic administration of psychostimulant drugs

leads to a significant changes in the dopaminergic system.

In humans, imaging studies found reduced dopaminergic function in cocaine-

addicted (Wu et al., 1997; Volkow et al., 1999). In animals studies, data from intense

continuous self-administration or repeated binge experimenter administered cocaine also

show significantly decreased extracellular basal DA (Weiss et al., 1992; Kreek and Koob,

8

1998). Although, as observed during an acute binge pattern of cocaine administration, the

extracellular DA levels rise following each dose of cocaine with much greater intensity in

the Nac than in the dorsal striatum, after chronic binge pattern cocaine, there are a

significant decrease in the extracellular DA levels at the basal time points, and a lower

elevation following each cocaine administration, both in the dorsal striatum and in Nac

(Maisonneuve et al., 1995). However, chronic administration of cocaine under intermittent

schedules rather than a binge-like or continuous pattern results in a rise in the

extracellular DA increase with repeated administration (Kalivas and Duffy, 1993; Pierce

and Kalivas, 1997a). The data described above show that the response of dopaminergic

neurons depended on the doses and pattern of exposure (Koob and Moal, 2006).

Continuous access seems to be more likely to produce decreases in firing and

transmission, and limited or intermittent access are more likely to produce later increases

in firing and DA neurotransmission (Koob and Moal, 2006).

Decreases in both D1 and D2 receptors have been observed after long-term,

heavy exposure to passive administration or self-administration of cocaine in rats

(Tsukada et al., 1996; Maggos et al., 1998), nonhuman primates (Moore et al., 1998a, b)

and humans (Volkow et al., 1993).

In in vitro studies, cocaine administration did not alter quantitatively measured DA

transporter (DAT) mRNA levels in the SN or the VTA following subacute (3 days) binge,

chronic (14 days) binge or 10 days withdrawal from a chronic binge administration pattern

(Maggos et al., 1997). However, decreases in DAT function including both binding and

mRNA levels have been observed in selective brain regions, following 10 days of

withdrawal from chronic cocaine administration (Kuhar and Pilotte, 1996).

The changes in brain dopaminergic function are likely to result in decreased

sensitivity to natural reinforcers since DA also mediates the reinforcing effects of natural

reinforcers, and on disruption of frontal cortical functions, such as the inhibitory control

and salience attribution (Volkow et al., 2003). These alterations could contribute to the

loss of control and compulsive drug intake that characterize addiction (Volkow et al.,

2003). Moreover, reduced DA-mediated reward could explain the high rates of

depression, irritability, anxiety, and suicide that have been reported in cocaine-addicted

individuals (Dackis and O'Brien, 2001).

9

1.4.2. Effects of cocaine upon the serotonergic system

Although the dopaminergic system still plays an important role in the investigation

of the behavioural effects of cocaine, it has became evident that models that involve only

DA do not fully explain the complex effects of cocaine on behaviour. The essential role of

the DAT in cocaine reinforcement has been challenged, namely with studies using

knockout mice, to show that mice lacking DAT still self-administer cocaine (Rocha et al.,

1998) and exhibit cocaine-conditioned place preference (Sora et al., 2001). However,

DAT/ Serotonin transporter (SERT) double knockout mice exhibit a lack of cocaine

reinforcement (Sora et al., 2001). Moreover, pretreatment with the 5-HT reuptake inhibitor

fluoxetine decreases intravenous cocaine self-administration (Carroll et al., 1990; Peltier

and Schenk, 1993). Thus, the focus of cocaine research expanded to include the

serotonergic system.

As already mentioned, serotonergic system is one of the main targets of cocaine in

brain. By binding to SERT, cocaine blocks 5-HT reuptake and, thus, causes an increase in

extracellular 5-HT levels (Ritz et al., 1990; Muller et al., 2003).

The increased concentration of 5-HT in the synaptic cleft causes profound

activation of 5-HT receptors. Currently, seven families of 5-HT receptors (5-HT1- 5-HT7)

with at least 14 subtypes are known to mediate the effects of 5-HT (Barnes and Sharp,

1999; Hoyer et al., 2002). In general, the serotonergic system in the brain regulates much

different behaviours, including locomotor activity, eating, drinking, and is deeply involved

in emotions and mood. Furthermore, 5-HT is an important factor in maintaining plasticity

at the cellular level, which is the basis of behavioural adaptation (Muller and Huston,

2006).

The contribution of the serotonergic system to the psychostimulant effects of

cocaine is demonstrated clearly by lesion studies and pharmacological manipulation of the

global activity of the serotonergic system in rodents and monkeys, and by transgenic

approaches in mice (Porrino et al., 1989; Loh and Roberts, 1990; Glatz et al., 2002; Muller

et al., 2003; Rothman et al., 2005; Wee et al., 2005). Several studies with selective 5-HT

receptor-ligands suggest the contribution of a large number of different 5-HT receptor

subtypes to the behavioural effects of cocaine (Muller and Huston, 2006). Stimulation of

serotonin-1A (5-HT1A) receptors facilitates the establishment of locomotor sensitization

(De La Garza and Cunningham, 2000; Carey et al., 2001). 5-HT1B receptor stimulation

appears to play a role in facilitating the reinforcing (Parsons et al., 1996b; Neumaier et al.,

2002) and discriminative properties of cocaine (Callahan and Cunningham, 1995). 5-HT2

receptor blockade attenuates cocaine-induced hyperactivity in mice (O'Neill et al., 1999;

10

Muller and Huston, 2006). While, 5-HT3 receptor antagonism have been shown to block

the behavioural expression of cocaine sensitization in rats and mice (Reith, 1990;

Kankaanpaa et al., 1996; King et al., 2000; Kankaanpaa et al., 2002).

Most of the 5-HT receptor subtypes that facilitate cocaine-induced behaviour are in

the VTA and the Nac, the core areas of the mesolimbic DA releasing system. In both

areas cocaine causes a profound increase in the extracellular concentration of the 5-HT

(Muller et al., 2003), which activates the local populations of 5-HT receptors. In fact,

several studies have shown that 5-HT receptors in these areas regulate the behavioural

effects of cocaine by modulating the dopaminergic response in the Nac (Kankaanpaa et

al., 2002; Szumlinski et al., 2004). For instance, microinfusion of 5-HT into the VTA

enhanced the release of DA in the Nac, with the same effect being observed with 5-HT3

receptor activation (Jiang et al., 1990; Chen et al., 1991; Van Bockstaele et al., 1994;

Matell and King, 1997).

Nevertheless, modulating only the serotonergic response also modulates the

behavioural effects of cocaine (Muller et al., 2002a) without influencing cocaine-induced

DA increase in the Nac (Muller et al., 2002b). Thus, there is evidence to support at least

two mechanisms by which the cocaine-induced increase in the extracellular 5-HT level

translates into behavioural changes: by direct stimulation of 5-HT receptors and by

indirect effects of 5-HT receptor stimulation that regulates the activity of other transmitter

systems (e.g. the dopaminergic system) (Muller and Huston, 2006).

Beside the role of the serotonergic system on psychostimulant action of cocaine,

long-term cocaine use induces adaptive processes in serotonergic neurons that contribute

to the emergence and persistence of a cocaine-dependent state. Withdrawal from

continuous access to cocaine self-administration was reported to decrease the

extracellular 5-HT in the Nac (Parsons et al., 1995, 1996a). This effect is consistent with

changes in 5-HT receptor function and transport which may contribute to a post-cocaine

deficiency in extracellular 5-HT (Koob and Moal, 1997; Kreek and Koob, 1998). Increased

5-HT1B mRNA in Nac shell and dorsal striatum (Hoplight et al., 2007) and increased

densities of 5-HT reuptake sites have been demonstrated in rats following repeated

cocaine exposure (Cunningham et al., 1992).

The serotonergic system has been implicated in the chronic effects of cocaine

(Pierce and Kalivas, 1997b) and alterations in 5-HT dynamics may be important in the

transition to addiction (Mash et al., 2000). In fact, many studies suggest that alterations in

serotonergic system contribute to the craving and depressed mood associated with

cocaine withdrawal (Pollack and Rosenbaum, 1991; Buydens-Branchey et al., 1997;

11

Buydens-Branchey et al., 1998; Harris et al., 2001). Moreover, these studies are

supported by the knowledge that 5-HT plays an important role in mood and impulse

control (Young et al., 1996; Lucki, 1998; Merens et al., 2007) and dysfunction in the

serotonergic neurotransmission has been linked to the etiology of a variety of clinical

disorders such as depression, anxiety and obsessive-compulsion disorder (Reilly et al.,

1997; Lucki, 1998).

1.5. Cocaine and the Neuroendocrine System

Cocaine’s interactions with the HPA (Hypothalamus-Pituitary-Adrenal) and HPG

(Hypothalamus-Pituitary-Gonads) axes system are not yet full understood. However,

cocaine related changes in these hormones have broad implications for normal

reproductive function as well as immune function (Mello and Mendelson, 1997). In

addiction, there is evidence that cocaine’s perturbation of the HPA axis may be related to

its reinforcing properties (Mello and Mendelson, 1997). An improved understanding of the

interactions between the neuroendocrine system and cocaine use and abuse will be

helpful in clarifying some aspects of the neurobiology of drug abuse.

1.5.1. The Hypothalamus-Pituitary-Adrenal axis

The HPA axis consists of a complex, well-regulated interaction between the brain,

anterior pituitary and adrenal cortex, and is the hormonal system that activates the

integrative physiological response to stress, which helps the organism to adapt to

increased demands and maintain homeostasis after challenge (Mello and Mendelson,

1997). This endocrine system is also vital for supporting normal physiological functioning,

regulates various body processes including digestion, the immune system, mood and

sexuality, and energy usage.

The synthesis and secretion of glucocorticoids (the final product of the HPA axis),

in the adrenal cortex is primarily mediated by the action of adrenocorticortipic hormone

(ACTH) on melanocortin-2 receptors integral to cortical cell membranes (Mountjoy et al.,

1992), with the adrenal sensitivity changing as a circadian function (Jasper and Engeland,

1994). Usually ACTH concentrations in the circulation are in the low picomolar range

(Dallman et al., 1987) and at these concentrations has a specific action in the adrenals,

but few extra-adrenal effects.

On the other hand, ACTH secretion from the anterior pituitary is usually entirely

controlled by the secretory activity of corticotrophin-releasing hormone (CRH) and

12

arginine vasopressin (AVP)- containing neurons in the medial parvocellular portion of the

paraventricular nuclei of the hypothalamus (PVN). Thus, circulating concentrations of

ACTH and glucocorticoids precisely reflect the prior secretion of CRF/AVP by the

hypothalamic median eminence (Alexander et al., 1996). Although CRH is required to

stimulate ACTH synthesis and secretion from the pituitary, AVP interacts with CRH to

potentiate the secretion of ACTH from the corticotrophs (Gillies et al., 1982; Antoni, 1993).

The CRH/AVP ratio in parvocellular neurons of the PVN changes under various conditions

and in this way can control the amount of ACTH released in response to stimulation of the

PVN (Antoni, 1993).

Figure 1.5.1.1- Schematic diagram of the hypothalamic-pituitary-adrenal axis. ACTH- adrenocorticotropic

hormone; Cort- corticosterone; CRH- corticotrophin-releasing hormone; (-), negative feed-back. Adapted from

The HPA axis function has a diurnal rhythm, for example, ACTH and glucocorticoid

levels are highest in the rat (a nocturnal species) in evening and lowest in the morning

(Dallman et al., 1987). These rhythms are generated and controlled by inputs from

suprachiasmatic nucleus to the CRH systems (Watts and Swanson, 1987, 1989) and

feedback actions of the circulating concentrations glucocorticoids on the HPA axis (Akana

et al., 1992a; Akana et al., 1992b). In fact, glucocorticoids are involved in the suppression

of their own release through negative feedback systems that inhibit the release of ACTH

(Keller-Wood and Dallman, 1984; Dallman et al., 1987). Feedback systems operate

primarily at the level of the parvocellular part of the PVN and anterior pituitary (Keller-

Wood and Dallman, 1984; Dallman et al., 1987; Cole et al., 2000), although other brain

13

sites such as the hippocampus and medial prefrontal cortex are also involved in the

regulation of HPA axis activity (Kovacs et al., 1986; Jacobson and Sapolsky, 1991; Diorio

et al., 1993).

In the CNS, corticosteroids exert their effects through two corticosteroids

receptors, a high-affinity low-capacity type I or mineralocorticoid receptors (MR) that are

saturated by low basal levels of the hormone, and a lower-affinity high-capacity type II or

glucocorticoid receptor (GR) that are activated by high glucocorticoid levels such as those

observed during the circadian peak or after a stressful event (Joels and de Kloet, 1994).

These cytosolic receptors regulate gene transcription when bound to DNA response

elements or by interactions with other transcription factors (Joels et al., 2004). The

distribution of MR is more discrete than that of GR. High expression of MR in

hippocampus has been shown in rats, humans and nonhuman primates (Krozowski and

Funder, 1983; Rupprecht et al., 1993; Sanchez et al., 2000). MR are also distributed in the

amygdala, prefrontal cortex, lateral septum, and in a variety of motor neurons in the brain

stem (Birmingham et al., 1984). In contrast to MR, GR are broadly distributed throughout

the brain and pituitary (Cintra et al., 1991). There is dense GR staining in hypothalamus,

limbic brain, cortex and brain stem. Of interest, most neurons that synthesize and secrete

monoamines contain GR (Fuxe et al., 1985; Harfstrand et al., 1986; Ceccatelli et al., 1989;

Cintra et al., 1991).

In the CNS, glucocorticoids influence protein synthesis, neuronal excitability and

neurotransmitter metabolism (McEwen, 1980; Keller-Wood and Dallman, 1984). In

addition, glucocorticoids are important regulators of brain development and neuronal

plasticity; they can influence virtually all aspects of neural development, including

neurogenesis, synaptogenesis, dendritic morphology and cell death (McEwen, 2000).

1.5.1.1. Effects of cocaine upon the HPA axis activity

There is increasing empirical support for the notion that dysregulation of the HPA

axis may increase vulnerability to depression, and a number of other psychiatric disorders,

as well as immune dysfunction and cardiovascular disease (Heinrichs et al., 1995;

Chrousos and Gold, 1998; Nemeroff, 1998). Cocaine, like stress, modulates HPA axis

activity and thereby contributes to the disruption of normal neuroendocrine function.

Acute cocaine produces a robust and dose-dependent activation of the HPA axis

in rats (Rivier and Vale, 1987; Saphier et al., 1993), rhesus monkeys (Sarnyai et al., 1996;

Broadbear et al., 1999b, a) and humans (Baumann et al., 1995b; Heesch et al., 1995).

Acute administration of cocaine stimulates ACTH and cortisol secretion in humans (Sholar

14

et al., 1998; Elman et al., 1999) and in rhesus monkeys (Sarnyai et al., 1996; Broadbear

et al., 1999a), and ACTH and corticosterone release in rats (Rivier and Vale, 1987;

Borowsky and Kuhn, 1991b; Levy et al., 1991).

There is considerable evidence that the stimulatory effects of cocaine on HPA axis

are mediated by CRH (Mello and Mendelson, 1997). Cocaine stimulates CRH release

from hypothalamic tissue in vitro (Calogero et al., 1989) and alters CRH levels in different

brain areas in vivo (Sarnyai et al., 1993). Moreover, cocaine-induced increases in ACTH

and corticosterone release are prevented by passive immunization against CRH (Rivier

and Vale, 1987; Sarnyai et al., 1992). These results show that the action of cocaine on the

HPA axis is mediated via CRH receptors and depends on the release of endogenous

hypothalamic CRH.

Although the exact mechanisms underlying the effects of cocaine on the HPA axis

remain to be clarified, it is increasingly apparent that cocaine-related stimulation of ACTH,

and by inference of CRH, is modulated by several interacting neurotransmitter systems

(Mello and Mendelson, 2002). Because CRH release is regulated, in part, by DA and 5-

HT, antagonists that are selective for DA or 5-HT receptors attenuate cocaine-induced

stimulation of ACTH (Borowsky and Kuhn, 1991b; Levy et al., 1991). Moreover, both DA

and 5-HT receptor agonists stimulate ACTH release in rats (Borowsky and Kuhn, 1991b;

Levy et al., 1994a; Baumann et al., 1995a)

In contrast to the acute stimulant effects of cocaine in the HPA axis, there are

conflicting reports in the literature as to whether chronic cocaine exhibits persistent

stimulatory effects on the HPA axis. Repeated cocaine exposure has been reported to

produce tolerance (Zhou et al., 1996), sensitization (Schmidt et al., 1995) or no changes

(Borowsky and Kuhn, 1991a; Levy et al., 1992) in the HPA axis response, depending on

the experimental design used (Mantsch et al., 2003). It is suggested that the frequency

and duration of cocaine administration, among other variables, may be of critical

importance in determining the effects of cocaine on HPA axis activity (Mantsch et al.,

2003).

Intermittently cocaine exposure usually does not alter basal levels of ACTH and

cortisol or corticosterone, or the hormonal response to acute stimulation with cocaine or

synthetic CRH factor (Borowsky and Kuhn, 1991a; Levy et al., 1992; Torres and Rivier,

1992a, b; Laviola et al., 1995; Broadbear et al., 1999b). Nevertheless, decreases in CRH

receptors have been reported after intermittently cocaine (Goeders et al., 1990; Mello and

Mendelson, 1997). Overall, knowledge obtained until nowadays seems to indicate that

15

intermittently cocaine exposure does not result in tolerance or sensitization of the HPA

axis to stimulation by cocaine or stress (Mello and Mendelson, 1997).

However, tolerance to the stimulatory effects of cocaine on HPA axis function has

been reported in rats following chronic binge pattern administration (Zhou et al., 1996).

This tolerance is observed as attenuated corticosterone and ACTH responses to cocaine

(Zhou et al., 1996; Zhou et al., 2003). In rats a significant reduction in plasma

corticosterone response to cocaine was observed on day 14 of binge administration as

compared with day 1 or 3 (Zhou et al., 1996). Moreover, the development of tolerance to

the effects of cocaine on the HPA hormone response is associated with a significant

reduction in the CRH mRNA levels in the hypothalamus (Zhou et al., 1996).

During early withdrawal from chronic binge cocaine administration, an activation of

the HPA axis of the rat has been observed, as indicated by the significant elevation of

plasma ACTH and corticosterone 1 and 4 days after withdrawal (Zhou et al., 2003),

however, 10 days after withdrawal, ACTH and corticosterone were at control levels (Zhou

et al., 2003). Increased basal levels of corticosterone 1 day after withdrawal from

intermittent cocaine administration has also been reported (Levy et al., 1994b). In

contrast, when rats are permitted to self-administer a very high dose of cocaine during

long-access sessions, plasma corticosterone is not elevated during acute withdrawal but

rather is decreased (Mantsch et al., 2000).

In humans, as already mentioned, cocaine also disrupts HPA axis function. Acute

cocaine administration increases plasma ACTH and cortisol levels in cocaine abusers

(Teoh et al., 1994; Baumann et al., 1995b; Elman et al., 1999). However, the effects of a

challenge dose of cocaine on ACTH secretion, are significantly lower in cocaine-

dependent men than in occasional cocaine users (Mendelson et al., 1998). Nevertheless,

one early study has shown higher basal plasma ACTH and cortisol levels in cocaine

addicts one day after the cessation of cocaine self-administration (Vescovi et al., 1992).

However, after a brief period of abstinence, basal and CRH stimulated ACTH and cortisol

levels in cocaine-dependent patients do not show differences from healthy subjects

(Vescovi et al., 1992; Baumann et al., 1995b; Mendelson et al., 1998; Jacobsen et al.,

2001a).

Behaviourally, cocaine use in humans has been reported to produce profound

subjective feeling of well being and a decrease in anxiety (Gawin and Ellinwood, 1989).

However, some of the major symptoms observed during withdrawal from chronic cocaine

intoxication can often included severe anxiety as well as restlessness, agitation and

depression (Gawin and Ellinwood, 1989). Interestingly, CRH has been reported to be

16

involved in a variety of neuropsychiatric disorders including depression and anxiety (Gold

et al., 1984; Musselman and Nemeroff, 1996), suggesting that the anxiety associated with

cocaine use and withdrawal may depend, in part, on the effects this psychostimulant on

the CRH release and subsequent activation of the HPA axis (Goeders, 1997). Anxiogenic-

like behaviour has also been observed in non-human animals using a variety of

behavioural paradigms, following acute, during chronic administration of cocaine and

during withdrawal (Costall et al., 1989; Fontana and Commissaris, 1989; Rogerio and

Takahashi, 1992; Sarnyai et al., 1995).

1.5.1.2. The HPA axis and the behavioural effects of cocaine

The HPA axis seems to have particularly importance in drug addiction (Piazza and

Le Moal, 1996; Goeders, 1997). The activity of the HPA axis prior to, at the time of, and

subsequent to cocaine availability appears to be an important determinant of whether or

not individuals will engage in cocaine-seeking behaviour (Goeders and Guerin, 1996b;

Mantsch et al., 1998; Mantsch and Goeders, 1999). The temporal concordance between

peak plasma cocaine levels, peak levels of ACTH, and reports of subjective high have

prompted speculation that perturbation of the HPA axis may contribute to cocaine’s

abuse-related effects (Goeders, 1997; Goeders, 2002b; Mendelson et al., 2002).

In fact, preclinical studies suggest that the reinforcing properties of cocaine may be

influenced by the HPA axis activity (Piazza and Le Moal, 1996; Goeders, 1997; Goeders,

2002a). Drug-naive rats that were surgically adrenalectomized did not learn to self-

administer cocaine (Goeders and Guerin, 1996b). Moreover, metyrapone, which blocks

corticosterone synthesis, significantly decreased cocaine-induced locomotion and relapse

of cocaine self-administration (Piazza et al., 1994; Goeders and Guerin, 1996a; Goeders

and Guerin, 1996b). Pretreatment with CRH receptor antagonist also produced dose-

related decreases in cocaine self-administration in rats (Goeders and Guerin, 2000). On

the other hand, elevated plasma corticosterone facilitates the initiation of cocaine self-

administration (Goeders and Guerin, 1996b; Mantsch et al., 1998). Dose-response studies

have shown that adrenalectomy induces a vertical downward shift to the dose-response

curve to cocaine during the maintenance phase (Deroche et al., 1997). These data

converge to suggest a role for the HPA axis in the modulation of cocaine reinforcement

(Goeders, 1997; Goeders, 2002a).

Although high levels of corticosterone are necessary to maintain self-

administration behaviour, drug-induced increases in hormone levels are not critical

(Marinelli and Piazza, 2002). Thus, self-administration of psychostimulants can

17

dramatically increase glucocorticoid secretion (Baumann et al., 1995b; Broadbear et al.,

1999b; Galici et al., 2000), but blocking this drug-induced increase in hormone levels does

not modify response to cocaine (Deroche et al., 1997). In addition, animals can

consistently self-administer cocaine at doses that do not increase hormone levels

(Broadbear et al., 1999a), further supporting the notion that drug-induced glucocorticoid

secretion is not essential to maintain self-administration behaviour (Marinelli and Piazza,

2002).

Although corticosterone plays a facilitatory role on many behavioural responses to

psychostimulants, its role in relapse is a little more controversial. Suppression of

glucocorticoids does not seem to have important effects on relapse induced by drug

priming. Thus, cocaine-induced reinstatement of cocaine self-administration is only

minimally decreased by adrenalectomy (Erb et al., 1998) and is not modified by

ketoconazole, which reduces circulating levels of corticosterone (Mantsch and Goeders,

1999). Instead, corticosterone may play a significant role in stress and cue-induced

reinstatement of drug seeking (Goeders, 2002a; Goeders and Clampitt, 2002).

1.5.1.2.1. Stress and increased vulnerability to cocaine

Scientists are ware of the existence of a complex relationship between stress, the

subsequent activation of the HPA axis and the neurobehavioural effects of cocaine for

many years now (Goeders, 1997; Piazza and Le Moal, 1998; Koob, 1999).

It has been hypothesized that exposure to physically or psychologically stressful

events may render an individual more sensitive to the reinforcing effects of drugs such as

cocaine (Piazza and Le Moal, 1996; Goeders, 1997; Piazza and Le Moal, 1998). This

hypothesis is supported by studies where prior exposure to stressful stimuli results in an

enhancement of behaviours pertaining to acquisition (Piazza et al., 1990; Haney et al.,

1995; Goeders and Guerin, 1996b), maintenance of cocaine self-administration (Miczek

and Mutschler, 1996), and reinstatement of cocaine-seeking in rats following extinction of

operant behaviour (Erb et al., 1998).

A faster acquisition of cocaine self-administration has been found in rats exposed

to social stress, such as exposure to a resident aggressive animal (Haney et al., 1995;

Tidey and Miczek, 1997). Physical stressors can also enhance the propensity to develop

cocaine self-administration. This has been shown for repeated tail pinch (Piazza et al.,

1990), food restriction (Papasava and Singer, 1985; Carr et al., 2000), and electric foot

shocks (Goeders and Guerin, 1996b; Xi et al., 2004). Very early life events, such as

prenatal stress, can also increase vulnerability to psychostimulants (Kippin et al., 2008).

18

The reinstatement of cocaine seeking behaviour can be elicited by exposure to

brief periods of intermittent electric footshock or food restriction stress in rats (Shaham et

al., 2000; Shalev et al., 2003). However, cocaine reinstatement was prevented in

adrenalectomized rats but not in adrenaloctomized animals with corticosterone

replacement. In addition, CRH receptor antagonists were also found to block (Erb et al.,

1998) or attenuate (Shaham et al., 1998) footshock-induced reinstatement in intact

animals and in adreneloctomized animals with corticosterone replacement (Erb et al.,

1998). This last example supports the view that CRH acting at extra-hypothalamic sites in

the brain also plays an important role in stress-induced reinstatement of cocaine seeking

(Shaham et al., 2000). In fact, acute cocaine has been reported to affect CRH-like

immunoreactivity in a number of brain regions that are not thought to be directly linked to

pituitary-adrenal activity, including basal forebrain and amygdala (Sarnyai et al., 1993;

Gardi et al., 1997). Extrahypothalamic CRH systems appear to be involved in certain

symptoms of acute drug withdrawal, such as anxiety, and in stress-induced relapse to

drug seeking (Sarnyai et al., 2001).

In contrast to the effects observed during acquisition or reinstatement, neither

exposure to footshock (Goeders and Guerin, 1996b) nor exogenous injections of

corticosterone affect ongoing cocaine self-administration. This inability to affect

maintenance of drug use is probably related to the fact that plasma corticosterone is

significantly elevated in a dose-related manner during cocaine self-administration, and

further increases in corticosterone are without effect, since a threshold critical for reward

has already been crossed (Goeders and Clampitt, 2002). However, low-dose cocaine self-

administration can be attenuated by drugs that inhibit the synthesis and/or release of

corticosterone, since in this case plasma concentrations of corticosterone is probably

reduce below the critical reward threshold (Goeders, 2002a).

In humans, there is also a growing literature that suggests a possible linkage

between stress and addiction. A number of studies have identified individuals with dual

diagnosis of post traumatic stress disorders (PTSD) and drug addiction (e.g. Zaslav,

1994; Donovan et al., 2001; reviewed by Jacobsen et al., 2001b). Although a causal

relationship between exposure to stress and drug addiction has not been clearly

established, prevalence estimates suggest that rates of drug abuse among individuals

with PTSD may be as high as 60-80%, suggesting relationship between stress and

increased drug addiction in some cases (Donovan et al., 2001).

Exposure to stress or simply the presentation of stress-related imagery have also

been identified as potent events for provoking relapse to drug seeking in humans (Lamon

and Alonzo, 1997; Sinha et al., 2000; Sinha, 2001). Clinical studies have demonstrated

19

that simple exposure to environmental stimuli or cues previously associated with drug

taking can produce intense drug craving (Robbins et al., 1999).

Preclinical investigations have also demonstrated cue-induced reinstatement (Meil

and See, 1996; See, 2002). Once again, several studies have suggested an important

role for the HPA axis in addiction, showing a link between HPA axis and the ability of

environmental cues to stimulate cocaine-seeking behaviour in rats. Pretreatment with, the

corticosterone synthesis inhibitor, ketoconazole, reversed the cue-induced reinstatement

of extinguished cocaine-seeking behaviour and also attenuated the conditioned increases

in plasma corticosterone observed during reinstatement (Goeders and Clampitt, 2002).

Pretreatment with a CRH receptor antagonist also resulted in a similar decrease in the

ability of environmental cues to stimulate cocaine-seeking behaviour (Goeders and

Clampitt, 2002).

It is not inherently intuitive how exposure to a stressor can increase vulnerability to

drug self-administration (Goeders and Clampitt, 2002). The preclinical literature suggests

that stress increases reinforcement associated with psychomotor stimulants, possibly

through a process similar to sensitization (Piazza and Le Moal, 1998; Goeders, 2002b),

whereby repeated intermittent injections of cocaine increase the behavioural and

neurochemical responses to subsequent exposure to the drug. Interestingly, exposure to

stressors or administration of corticosterone can also result in sensitization to the

behavioural and neurochemical (e.g., Nac DA) response to cocaine (Rouge-Pont et al.,

1995; Prasad et al., 1998). These effects are attenuated in adrenalectomized rats (Prasad

et al., 1998; Przegalinski et al., 2000) or when corticosterone synthesis is inhibited

(Rouge-Pont et al., 1995). The ability of stressors to facilitate the acquisition of drug self-

administration may, therefore, result from a similar sensitization phenomenon, perhaps

involving DA (Goeders, 1997; Piazza and Le Moal, 1998).

1.5.1.2.1.1. Interaction between stress, glucocorticoids and vulnerability to drugs

Stress, through activation of the HPA axis and the release of glucocorticoids,

influences various regions of the brain including dopaminergic neurons (Piazza and Le

Moal, 1996; Piazza and Le Moal, 1997; Piazza and Le Moal, 1998), that express

corticosteroid receptors (Harfstrand et al., 1986). In normal situations, glucocorticoids

state-dependently increase dopaminergic function, especially in mesocorticolimbic

regions, during various consummatory behaviours exhibited in rodent’s active period of

the light/dark cycle (Piazza and Le Moal, 1996). The interaction of glucocorticoids with

mesocorticolimbic dopaminergic system may have a significant impact on vulnerability to

20

self-administer psychostimulant drugs, since increased dopaminergic transmission in

these pathways is critical for the reinforcing properties of abusive drugs, rendering the

individuals more susceptible to the drug reinforcement (Roberts et al., 1980; Koob and

Moal, 1997).

GR have been identified in rodent brain on dopaminergic neurons in the VTA and

SN as well as in dopaminergic terminal regions including the Nac and prefrontal cortex

(Harfstrand et al., 1986; Diorio et al., 1993; Cintra et al., 1994). Stress-induced elevations

in corticosterone may play a critical role in activation of dopaminergic transmission, with

corticosterone treatment increasing, and adrenalectomy decreasing extracellular DA

levels in Nac and prefrontal cortex of rodents (Imperato et al., 1989; Piazza et al., 1996)

This suggest that stress-induced increases in glucocorticoids may interact with

mesocorticolimbic brain regions to facilitate drug taking behaviour .

1.5.2. The Hypothalamus-Pituitary-Gonadal axis

The HPG axis, also known as reproductive axis, is a critical part in the

development and regulation of the reproductive function.

1.5.2.1. The Hypothalamus-Pituitary-Testicular axis

Regulation of the hypothalamus-pituitary-testicular (HPT) axis begins with at the

level of the hypothalamus, where neurosecretory cells synthesize and release

gonadotropin-releasing hormone (GnRH) in a pulsatile fashion into the hypothalamic-

hypophysial-portal circulation. In response gonadotropes in the anterior pituitary

synthesize and release the gonadotropins follicle-stimulating hormone (FSH) and

luteinizing hormone (LH), both of which ultimately control gonadal function (Swerdloff et

al., 2002).

In males, LH is the primary stimulus for the secretion of testosterone and FSH

stimulates spermatogenesis (Swerdloff et al., 2002). Testosterone is secreted by the

testicular Leydig cells, but only when they are stimulated by LH from the anterior pituitary

gland (Swerdloff et al., 2002).

FSH seems to mediate its effects on the testes predominantly through its action on

the Sertoli cells. FSH is known to bind a specific receptor on these cells and stimulate the

production of a large number of proteins including the inhibin-related peptides, androgen-

binding protein, transferring, androgen receptor, and γ-glutamyl transpeptidase (Swerdloff

et al., 2002).

21

Figure1.5.2.1.1- Schematic diagram of the male hypothalamic-pituitary-testicular axis. E2- estradiol; FSH-

follicle-stimulating hormone; GnRH- gonadrotopin-releasing hormone; LH- luteinizing hormone; T-

testosterone; (-)- negative feed-back. Adapted from (Norman and Litwack, 1998).

Beside the stimulation by the GnRH, gonadal steroids and testicular peptide

substances, such as activin and inhibin action on the gonadotropes provides additional

modulatory effects (Swerdloff et al., 2002). The primary regulator of LH secretion is the

serum level of testosterone (Swerdloff et al., 2002). It appears that the inhibitory effects

are largely accomplished at both the hypothalamic and pituitary levels (Swerdloff et al.,

2002).

Several inhibitory peptides (e.g., activin and follistatin) have been postulated as

selecting FSH inhibitors, but evidence to relate changing serum levels of either peptide

with increased FSH have not been shown in men. Thus, the understanding of FSH

regulation is incomplete (Swerdloff et al., 2002).

The secretion of GnRH is regulated in a complex fashion by neuronal input from

higher cognitive and sensory centers and by the circulating levels of sex steroids and

peptide hormones such as prolactin, activin, inhibin and leptin (Swerdloff et al., 2002). The

local effectors of GnRH synthesis and release include a number of neuropeptides,

opioids, catecholamines, indolamines, nitric oxide and excitatory amino acids, γ-

aminobutyric acid (GABA), neuropeptide Y, vasoactive intestinal peptide, and CRH. In

general, the catecholamines, excitatory amino acids, and nitric oxide in physiologic

22

amounts are stimulatory, whereas opioid peptides and β-endorphin are inhibitory

(Swerdloff et al., 2002). Testosterone, either directly or through its metabolic products,

estradiol and dihydrotestosterone (DHT) has predominantly inhibitory effects on the

secretion and release of GnRH (Swerdloff et al., 2002).

1.5.2.2. The Hypothalamus-Pituitary-Ovaries axis

The female hormonal system, like that of the male, consists of three hierarchies of

hormones, as follows: A hypothalamic releasing hormone, the GnRH, that like in males

stimulates the anterior pituitary to release FSH and LH, that in turn will stimulate the

ovarian to produce estrogens and progestins (Guyton and Hall, 2006).

Figure 1.5.2.2.1- Schematic diagram of the female hypothalamic-pituitary-ovarian axis. Depending upon

hormone concentration, time course, and prior hormonal status, E2 can either inhibit or stimulate LH secretion

through effects at either the hypothalamus or pituitary. E2- estradiol; FSH- follicle-stimulating hormone; GnRH-

gonadrotopin-releasing hormone; LH- luteinizing hormone; P- progesterone; (-)- negative feed-back; (+),-

positive feedback. Adapted from (Norman and Litwack, 1998).

By far the most important of the estrogens is estradiol, and the most important

progestin is progesterone (Guyton and Hall, 2006). Estradiol is produced by the ovarian

follicles, and progesterone is secreted from the ovarian follicle and from corpus luteum

after ovulation. These hormones are not secreted in constant amounts throughout the

female monthly sexual cycle, but drastically differing rates during different parts of the

cycle (Guyton and Hall, 2006). In fact, this changes in levels of gonadotropins and

gonadal steroid hormones define the functional distinct phases of the monthly sexual

23

cycle: the follicular phase, the ovulatory phase, and the luteal phase (Guyton and Hall,

2006).

Figure 1.5.2.2.2- Schematic diagram of the pattern of changes in the gonadotropins (LH and FSH) and the

ovarian steroids hormones (estradiol and progesterone) across a typically menstrual cycle in human females.

Adapted from (Guyton and Hall, 2006).

During each month of the female sexual cycle, there is a cyclical increase and

decrease of both FSH and LH. During the first few days of each monthly female sexual

cycle, the concentrations of both FSH and LH secreted by the anterior pituitary gland

increase slightly to moderately, with a increase in FSH slightly greater than that of LH and

preceding it in a few days. Adequate FSH levels are necessary for normal development

and maturation of the ovarian follicles (Goodman and Hodgen, 1983) and the early growth

of the primary follicle is stimulated mainly by FSH alone, LH is necessary for final follicular

growth and ovulation (Guyton and Hall, 2006). About 2 days before ovulation (for reasons

that are not completely understood) the rate of secretion of LH by the anterior pituitary

gland increases markedly, FSH also increases at the same time, and the FSH and LH act

synergistically to cause rapid swelling of the follicle during the last few days before

ovulation, leading to decrease in the rate of secretion of estrogen and a increase in

amounts of secretion of progesterone (Guyton and Hall, 2006). During the periovulatory

phase, the preovulatory LH and FSH surge is stimulated by increases in estradiol levels,

and an abrupt increase in progesterone precedes the onset of the LH surge. LH levels

decrease after ovulation and the luteal phase of the menstrual cycle begins (Nippoldt et

al., 1989). Progesterone levels gradually increase and remain elevated until the

24

postovulatory corpus luteum regresses. Throughout the luteal phase the levels of FSH are

low.

Gonadotropin and ovarian steroid hormone levels are controlled by a complex and

changing pattern of reciprocal stimulation and inhibition across the monthly sexual cycle

(Nippoldt et al., 1989). Estrogen in small amounts has a strong effect to inhibit the

production of both LH and FSH. Also, when progesterone is available, the inhibitory effect

of estrogen is multiplied, even though progesterone by itself has little effect (Nippoldt et

al., 1989). In addition to the feedback effects of estrogen and progesterone, other

hormones seem to be involved, especially inhibin. This hormone inhibits the secretion of

FSH and, to a lesser extent, the secretion of LH by the anterior pituitary gland (Guyton

and Hall, 2006).

Multiple neuronal centers in the higher brain’s limbic system transmit signals into

the arcuate nuclei to modify both the intensity of GnRH release and the frequency of the

pulses (Guyton and Hall, 2006). It has been suggested that gonadal steroids as well as

opioid peptides are important in the inhibitory regulation of GnRH release (Yen et al.,

1985). In addiction, the system is modulated further in a complex fashion by the combined

action of inhibin, activin, and follistatin (Norman and Litwack, 1998).

1.5.2.2.1. Estrous cycle

Although the estrous cycle of the rat is shorter than the reproductive cycle of

humans (4 vs. 28-35 days), the endocrinological changes closely resemble those seen in

humans (Quinones-Jenab et al., 2001). The 4-5 day cycle is composed of proestrus,

estrus, metestrus and diestrus (Barbacka-Surowiak et al., 2003).

Ovarian follicles and oocytes grow, mature and differentiate to reach the ability to

ovulate during estrus, these processes are controlled first by FSH and then by LH

(Barbacka-Surowiak et al., 2003). Gradually increasing FSH level stimulates the

proliferation of follicular cells and synthesis of estradiol, the LH determines the

differentiation of follicles (Barbacka-Surowiak et al., 2003). The maximum level of estradiol

turns on a positive feedback mechanism resulting in the preovulatory LH surge (Chappell

et al., 2000), the LH surge usually occurs on the day of proestrus and later during

proestrus, the preovulatory peak level of progesterone occurs (Barbacka-Surowiak et al.,

2003). Estrus is the time of ovulation, mating and oocyte fertilization, while a short

postovulatory metestrus is the time of corpus luteum formation. Diestrus can be

significantly shortened or even skipped during increased reproductive activity in spring,

whilst it can be markedly prolonged in late fall or winter. Diestrus is characterized by the

25

onset of follicular activity and estrogen surges, while progesterone levels remain low

(Barbacka-Surowiak et al., 2003).

Figure 1.5.2.2.1.1- Relative concentrations of ovarian hormones during the rat estrous cycle. Estrogen and progesterone fluctuate during the rat estrous cycle. In diestrus, estrogen levels begin to rise and

progesterone levels are low. In proestrus, estrogen levels peak and descend while progesterone levels rapidly

peak and decline by the end of the phase. Estrogen and progesterone decline during estrus and begin to rise

by the end of metestrus. Adapted from (Quinones-Jenab et al., 2001).

1.5.2.3 Effects of cocaine on the HPG axis

A number of studies have demonstrated cocaine’s modulation of the HPG axis.

Clinical studies indicate that women who abuse cocaine may have a variety of menstrual

cycle disorders, including amenorrhea and luteal phase dysfunction, which compromise

fertility (Smith et al., 1984; Mello and Mendelson, 1997). However, clinical evidence of

cocaine’s disruptive effects on reproductive function is often complicated by the fact that

many human cocaine abusers are polydrug abusers (Teoh et al., 1992; Mello and

Mendelson, 1997). Nevertheless, cocaine effects on different components of the

reproductive cycle have been demonstrated by preclinical studies. Chronic cocaine self-

administration and cocaine withdrawal were both associated with disruptions of menstrual

cycle regularity compared with control conditions in rhesus monkeys (Mello et al., 1997).

And chronic exposure to high doses of cocaine (10-20 mg/kg/day) disrupted estrous

cyclicity in rats and decreased rates of ovulation (King et al., 1990; King et al., 1993). In

the case of the estrous cycle it was demonstrated that the disruption of the cyclicity

depends on the cocaine treatment regimen (Booze et al., 1999; Sell et al., 2000), namely

it was found to be cocaine dose-dependent (King et al., 1993).

26

The mechanisms through which cocaine disrupts reproductive function are poorly

understood (Mello et al., 1997). It may be through cocaine’s direct stimulation of LH

release, or indirect through putative changes in ovarian hormone concentrations that

could alter feedback control of gonadotropin release (Mello et al., 1990; Mello et al.,

1993). In fact, acute administration of cocaine changes basal levels of anterior pituitary

hormones that are important for reproductive cycle normalcy. Frequent repetition of these

acute hormonal effects of cocaine might contribute to the reproductive cycle abnormalities

observed during chronic cocaine self-administration (Mello et al., 1997).

Acute administration of cocaine stimulates the release of LH in men and women

(Mendelson et al., 1989; Heesch et al., 1996; Mendelson et al., 2001; Mendelson et al.,

2002), as well as in male and female rhesus monkeys (Mello et al., 1990). This effect was

not predictable from the pharmacological actions as an indirect DA agonist of cocaine or

from the clinical literature on DA-gonadotropin interactions (Judd et al., 1979; Mutiara et

al., 2006). It has been suggested that cocaine’s stimulation of LH reflects a cocaine

action upon the GnRH release and not a direct action on the pituitary (Mello and

Mendelson, 1997; King et al., 2001). Acute administration also increases FSH in men

(Heesch et al., 1996). Acute cocaine increases plasma estradiol levels in female rhesus

monkeys during the follicular phase but has no effects on estradiol levels during the luteal

phase (Mello et al., 2000) and do not alter progesterone levels during either the follicular

or the luteal phase of the menstrual cycle (Mello et al., 2000). Acute cocaine treatment

also did not alter serum levels of testosterone in men (Mendelson et al., 2003). In addition,

in men, chronic cocaine abuse had no demonstrable effects on pulsatile release patterns

of LH or testosterone (Mendelson et al., 1989).

In rodents, acute cocaine transiently increased LH, but not FSH, in female rats

(King et al., 2001). However, in contrast to acute stimulatory effects of cocaine on LH, this

hormone levels measure at proestrus and diestrus were significantly lower after chronic

high-dose cocaine treatment (King et al., 1990). Progesterone plasma levels were

increased after acute and chronic cocaine administration in males and female rats

(Quinones-Jenab et al., 2000a; Walker et al., 2001a; Chin et al., 2002). In female rats, the

cocaine-induced levels of progesterone fluctuated within the estrous cycle and reached

their highest levels during proestrus (Quinones-Jenab et al., 2000b). Acute cocaine

administration did not affect plasma levels of estrogen, however, cocaine-stimulated

progesterone release was greatest when estrogen levels were at their highest (Walker et

al., 2001a). Cocaine has been shown to decrease testosterone levels in male rats after

acute and chronic treatment (Chin et al., 2002; Festa et al., 2003), whereas others

27

showed no changes in testosterone levels after an acute cocaine injection (Walker et al.,

2001b).

As described earlier, the normal reproductive function depends on the correct

interaction between GnRH, anterior pituitary and gonadal steroids hormones, thus

cocaine-induced alterations in the HPG axis seems to be, at least, one of the causes for

the clinical evidences of cocaine’s disruptive effects on the reproductive function. For

example, although the functional implications of a cocaine-related increase in LH are

unclear, repeated episodes of cocaine intoxication accompanied by elevated LH levels

could compromise folliculogenesis and prompt early ovulation (Zeleznik, 1981; Dierschke

et al., 1985; Mello and Mendelson, 1997). On the other hand, due to the profound effects

of the gonadal hormones on the modulation of CNS plasticity, cocaine-induced alterations

in the HPG axis may play a key role in modulating CNS neuronal functions and thus be an

important component in the cascade of events that follows the administration of cocaine

(Quinones-Jenab et al., 2001).

1.5.2.4. Effects of sex and sex hormones in the response to cocaine

Clinical and preclinical studies have invalidated the long-standing view of cocaine

as a predominately male problem. The current literature suggests that women are more

sensitive than men to the addictive properties of cocaine and could be more vulnerable to

its powerful psychostimulanting effects (Kosten et al., 1993; Chen and Kandel, 2002).

More severe cocaine use patterns are observed in females at the time of intake (Robbins

et al., 1999). Moreover, women consume cocaine by more addictive routes and progress

to dependence more rapidly than men (McCance-Katz et al., 1999). In fact, prevalence of

cocaine addiction was actually shown to be higher in adolescent females than adolescent

males (Kandel et al., 1997). Women also report shorter abstinence periods between

cocaine use than men (Kosten et al., 1996) together with stronger cravings in response to

cocaine cues as compared to men (Robbins et al., 1999).

It is not likely that these effects in women are due to differences in cocaine

metabolism as it has been demonstrated that blood levels of cocaine, after intravenous

administration are comparable in men and women, and do not vary significantly across

the menstrual cycle (Mendelson et al., 1999a). Also, no sex differences were found in

cocaine pharmacokinetics after intranasal cocaine administration (McCance-Katz et al.,

2005). Similarly, in rhesus monkeys, cocaine pharmacokinetics are similar in males and

females after intravenous administration (Mendelson et al., 1999b).

28

Preclinical data also has demonstrated sex differences in cocaine response. For

example, female rats do acquire cocaine self-administration more rapidly and at lower

cocaine doses than males, and after acquisition female rats also self-administer more

cocaine than did males (Lynch and Carroll, 1999; Elman et al., 2001), indicating that

females are more sensitive to the reinforcing effects of cocaine (Russo et al., 2003a).

Reinstatement of extinguished cocaine-reinforced responding is also greater in female

rats (Lynch and Carroll, 2000). Adding to that, female rats exhibit greater locomotor and

stereotypic behaviour than male rats after acute cocaine administration (Sell et al., 2000;

Chin et al., 2002; Festa et al., 2004). Regardless of dose, injection frequency, or length of

cocaine administration, female rats consistently demonstrate behavioural sensitization

after fewer cocaine injections as compared to male rats (Glick et al., 1983; Chin et al.,

2002). However, no sex differences in behaviour stereotypy were observed after chronic

cocaine administration, thus suggesting that locomotor and motor stereotypy respond

differentially to chronic cocaine (Chin et al., 2002).

The dynamic endocrinological profile of females has also been shown to affect

cocaine-induced responses in both clinical and preclinical studies.

In a human study, female cocaine users in the luteal phase had an attenuated

subjective response to smoked cocaine and less desire to smoke cocaine than did those

in the follicular phase (Sofuoglu et al., 1999). Also, a greater “high or good feeling” was

reported during the follicular phase after cocaine intake (Evans et al., 2002). Because the

luteal phase of the cycle is characterized by high levels of progesterone, these studies

suggest that progesterone attenuates the subjective effects of cocaine (Festa and

Quinones-Jenab, 2004).

Several preclinical studies have shown that the estrous cycle also influence the

behavioural response to acute cocaine administration in rats. Rats in estrus demonstrate

a higher incidence of cocaine-induced stereotypic and locomotor activity than do rats

during diestrus (Quinones-Jenab et al., 1999; Sell et al., 2000). Unlike studies of acute

cocaine administration, results from studies examining estrous cycle effects during chronic

cocaine administration have not yielded consistent results (Festa and Quinones-Jenab,

2004).

A clear picture is emerging which suggests that the biological basis of sex-specific

differences in cocaine effects resides in the disparate regulation of the CNS by male and

female gonadal hormones (Quinones-Jenab et al., 2001; Lynch et al., 2002).

For instance, gonadectomy of female rats decreased locomotor activity in

response to acute cocaine (Chin et al., 2002). However, gonadectomized female rats

29

treated with estrogen, or estrogen and progesterone showed enhanced locomotor activity

responsiveness to cocaine when they were compared to gonadectomized female rats

treated with progesterone alone or vehicle (Sell et al., 2000). Also chemical blockade or

surgical removal of estrogen do reduce cocaine self-administration during acquisition,

when compared to estrogen producing rats (Lynch et al., 2001).

Gonadectomy of male rats either increase or have no effect on behavioural

responses to acute cocaine (van Luijtelaar et al., 1996; Walker et al., 2001b; Chin et al.,

2002). Discrepancies in the results may be due to the cocaine dose or the strain of rat

used. Chen et al. (2003) showed that in gonadectomized male rats testosterone

replacement attenuated cocaine-induced behavioural stereotypy and locomotion. It was

suggested that testosterone reduced cocaine-induced sensitization of behavioural

stereotypy by increasing reuptake of striatal DA (Chen et al., 2003).

Overall, acute and chronic cocaine administration studies have demonstrated that

estrogen increases, testosterone reduces, and progesterone reduces or has no effect on

cocaine-induced activity in rodents (Festa and Quinones-Jenab, 2004). Although the

number of clinical studies is limited, the inhibitory effects of progesterone have been

consistent in both clinical and rodent studies (Festa and Quinones-Jenab, 2004).

Given the increasing evidence that there are sex differences in response to

cocaine, and that these differences might be related to fluctuations in gonadal hormone

levels, studies have attempted to determine whether there are pharmacokinetic

differences between males and females and whether hormonal fluctuations in females

alter the pharmacokinetics of cocaine. Most studies in rodents have reported no

differences in cocaine plasma levels between males and females (Bowman et al., 1999;

Festa et al., 2004). Further, while some studies have shown sex differences in cocaine

metabolite levels, the results have been inconsistent (Bowman et al., 1999; Chin et al.,

2001; Festa et al., 2004). Moreover, based on the data available in both monkeys and

humans, differences in the pharmacokinetics of cocaine across the menstrual cycle or

between males and females appear to be minimal (Kosten et al., 1996; Mendelson et al.,

1999a; Sofuoglu et al., 1999; Evans et al., 2002; Evans and Foltin, 2006). Thus, the

evidence that females and male have comparable cocaine levels in both plasma and brain

suggests that reported sex differences in the response to cocaine are not due to

substantial differences in brain drug levels (Bowman et al., 1999).

Still, there is considerable preclinical evidence that sex hormones modulate the

dopaminergic transmission. For instance, the rates of release and uptake of DA in the

dorsal striatum are greater in females than in males and the density of striatal DA

30

receptors is higher in females (Andersen and Teicher, 2000; Walker et al., 2000).

Moreover, dopaminergic neurotransmission varies throughout the estrous cycle,

demonstrating that dopaminergic activity is sensitive to endogenous fluctuations of

ovarian hormones (Davis et al., 1977; Becker and Ramirez, 1981; Thompson and Moss,

1994).

Ovarian hormones modulate the dopaminergic system at different levels. Estrogen

acts directly on the striatum to affect DA release (Becker, 1990a), turnover rates (Becker,

1990b) and reuptake (Thompson et al., 2000). Additionally, direct infusions of estrogen

into Nac result in increased DA levels that are significantly higher than those observed

following vehicle administration (Thompson and Moss, 1994). Mesocorticolimbic DA

release and reuptake are also controlled by progesterone (Cabrera et al., 1993). However,

the modulation by progesterone seems to be bimodal, where according to progesterone

plasma levels, DA release is either synergized or inhibited (Cabrera et al., 1993). Similarly

to the dopaminergic system, estrogen and progesterone can alter the function of the

serotonergic neuronal system at various levels, including secretion, receptor binding sites

and transporters (Luine, 1993; Bethea et al., 1998; Flugge et al., 1999). In male rats,

testosterone enhances turnover of both DA and 5-HT and also affects 5-HT binding sites

as well as uptake sites (McQueen et al., 1999).

The data presented above seems to indicate that sex hormones may influence

cocaine reward by altering monoamine neurotransmitter systems in the striatum and

mesolimbic areas (Becker, 1999; Russo et al., 2003b).

Given that cocaine remains a major public health problem and appears to be

increasing among women, understanding the interaction between cocaine, hormones and

behaviour will facilitate the development of effective treatment strategies for cocaine

abusers that may need to be gender-related.

1.5.3. Interactions HPG/ HPA axes and the response to cocaine

Countless studies have shown that stress and HPA axis activation inhibits

reproductive function and behaviour. The first line of evidence derives from the

observation that the administration of CRH results in an immediate decrease in pulsatile

GnRH and LH release (Olster and Ferin, 1987; Petraglia et al., 1987). The suppression of

the GnRH pulse generator by a variety of stressful stimuli can be blocked by CRH

antagonists (Rivier et al., 1986; Maeda et al., 1994). The suppressive effects of the HPA

system on the HPG function are also manifested at least in part as an inhibition of

gonadotropin release through effects of glucocorticoid hormones on the hypothalamus

31

and/or anterior pituitary gland. Treatment of adult male rats with corticosterone decreases

GnRH mRNA levels (Gore et al., 2006). Moreover, glucocorticoids suppress GnRH

release both in hypothalamic tissue in vitro (Calogero et al., 1999) and in GT1-7 cells

(Attardi et al., 1997). Decreases in circulating LH levels may also be due, at least in part,

to direct actions of corticosterone on pituitary gonadotropes. These cells have been

shown to express GRs in several studies (Kononen et al., 1993; Teitsma et al., 1999;

Breen et al., 2004), and they are also likely to be influenced by other pituitary cells

expressing GRs (e.g. corticotropes). In addition, the pituitary GnRH receptor may be

involved, as glucocorticoids can reduce GnRH-stimulated LH release directly from

pituitary cells (Briski and Sylvester, 1991).

There is also evidence that stress-activated inputs, including catecholaminergic

inputs to the hypothalamic PVN suppress LH pulses via CRH (Maeda et al., 1994).

In addition to the direct and indirect effects of cocaine on gonadotropins, cocaine

abuse may also disrupt the reproductive function by stimulation of the HPA axis. It is

possible that the acute stimulatory effects of cocaine on the HPA axis contribute to the

reproductive dysfunction observed during cocaine abuse (Mello et al., 1997).

This relationship between HPA and HPG axes is by no means unidirectional. The

reproductive status and gender impact heavily on both basal and stress-induced HPA axis

activity (Dallman et al., 2002).

Adult female rats have higher basal and stress levels of ACTH and corticosterone

than do males (Lesniewska et al., 1990b). The higher basal levels of corticosterone in

females are partly buffered by their higher levels of corticosteroid binding globulin

(McCormick et al., 2002) as only unbound corticosterone is generally considered to be

biologically active (Rosner, 1990). However, females have higher levels of free

corticosterone in response to stressors (Lesniewska et al., 1990b; Tinnikov, 1999; Beiko

et al., 2004). Females also have higher levels of CRH (Hiroshige et al., 1973) and

increased levels of CRH mRNA in the PVN (Watts and Swanson, 1989).

The sex differences in HPA axis function are largely due to sex hormonal

regulation of the HPA axis (Kudielka and Kirschbaum, 2005; McCormick and Mathews,

2007). Female rats have higher CRH, ACTH and corticosterone levels during proestrus,

when estradiol levels are high, than during other phases of the estrous cycle (Hiroshige et

al., 1973; Atkinson and Waddell, 1997; Walker et al., 2001a). Furthermore, plasma ACTH

and corticosterone levels in response to stress are higher during proestrus than during

other phases of the cycle (Viau and Meaney, 1991; Carey et al., 1995). Gonadectomy of

adult female rats reduces ACTH and corticosterone levels (Viau and Meaney, 1991;

32

McCormick et al., 2002; Seale et al., 2004b). Animal studies consistently reveal a strong

stimulatory influence of estradiol on HPA axis function (Lesniewska et al., 1990b; Viau

and Meaney, 1991; Norman et al., 1992; McCormick and Mathews, 2007), with

modulatory effects on MR and GR (Burgess and Handa, 1992; Carey et al., 1995).

Estradiol decreases GR binding and mRNA levels in various brain areas (Burgess and

Handa, 1992; Carey et al., 1995). Moreover, estradiol may directly enhance CRH gene

transcription in the hypothalamus through binding to estrogen-responsive elements on the

CRH gene transcription (Vamvakopoulos and Chrousos, 1993). Estradiol also directly

stimulate corticosterone secretion in dispersed adrenocortical cells (Nowak et al., 1995).

Thus, estrogen may influence adrenal corticosterone secretion directly and indirectly.

Although estradiol-mediated effects seem to be the most potent modulators of

HPA axis, stress regulation by progesterone might also contribute. In animal studies, it

was showed that progesterone can function as a glucocorticoid antagonist; can bind to

GR and MR receptors; can increase the rate of dissociation of glucocorticoids from the

receptor; and can diminish the effectiveness of corticosterone feedback on stress

responsiveness (Ahima et al., 1992; Carey et al., 1995).

Androgens exert specific and in part contrasting effects (inhibitory) at different

levels of HPA axis regulation as shown in several animal studies (Bingaman et al., 1994;

Handa et al., 1994; Viau and Meaney, 1996). Studies on gonadectomized males as adults

submitted to androgen replacement indicate that androgens typically inhibit the HPA axis

response to stress (Handa et al., 1994; Seale et al., 2004a), although there are some

reports of no effect of adult male gonadectomy on corticosterone levels (Lesniewska et

al., 1990b; Lesniewska et al., 1990a; Chen and Herbert, 1995).

Testosterone influences adrenal steroidogenesis leading to decreased

corticosterone levels in the adrenal gland (Malendowicz and Mlynarczyk, 1982). At central

levels androgen inhibits hypothalamic CRH (Bingaman et al., 1994), and replacement of

androgens limited to the medial preoptic area decrease corticosterone release in

response to stress in gonadectomized rats, while increasing GR levels (Viau and Meaney,

1996; McCormick et al., 2002). It also should be noted that testosterone can also exert

estrogenic effects after being metabolized to estrogen by aromatization in both brain and

peripheral tissues (Finkelstein et al., 1991; Bagatell et al., 1994).

Cocaine, like other stressors, is known to exert sexual-dimorphic HPA axis effects

in rats. Female have greater ACTH and corticosterone plasma levels after acute cocaine

exposure than do males (Kuhn and Francis, 1997). Moreover, the HPA axis response to

cocaine is strongly influenced by activational effects of ovarian hormones (Walker et al.,

33

2001a). Gonadectomy decreased cocaine-induced ACTH and corticosterone plasma

levels response in females, whereas castration had no effect on males (Kuhn and Francis,

1997). The estrous cycle stage has also been shown to influence cocaine-induced

secretion of ACTH, that are greatest in rats in proestrus (Walker et al., 2001a). The

variation in cocaine-stimulated HPA axis activation with the estrous cycle stage and

gonadectomy, support a role for ovarian steroids in the increased HPA axis responses of

females to cocaine (Walker et al., 2001a). This sexually dimorphic HPA axis responses to

cocaine may mediate some aspects of behavioural responses to cocaine including

cocaine self-administration (Walker et al., 2001a).

1.6. The adolescence

Adolescence is defined operationally as the transition period that characterizes the

passage from childhood to adulthood (Spear, 2000). This period encompasses not only

reproductive maturation, but also cognitive, emotional and social maturation (Sisk and

Foster, 2004).

The adolescent brain is a brain in transition, and differs anatomically and

neurochemically from that of the adult (Spear, 2000). This remodeling of brain appears

highly conserved across species and promotes behaviours supporting the evolutionary

fitness theme of optimal species reproduction (Ernst and Mueller, 2008). Adolescence is

likewise characterized by expression of a number of typical behavioural features, such as

increased novelty seeking and risk taking (Irwin, 1989), and a shift in social affiliation

towards more peer-based social interactions (La Greca et al., 2001). This is the time when

individuals begin to move away from the familiar cell and acquire independence in order to

fulfill adult roles in terms of reproductively mature and socially competent behaviour. This

impetus to move away from the protective nest is necessary for preventing genetic

inbreeding and for enabling genetic diversity, which serves the overall evolutionary fitness

goal (Ernst and Mueller, 2008). Behaviours of novelty-seeking and risk-taking facilitate this

transition (Spear, 2000; Ernst and Mueller, 2008). In the human adolescent, these

propensities may be expressed however, in drug use, as well as a variety of other deviant

behaviours (Spear, 2000).

Observation suggests enhanced mood liability and emotional intensity during

adolescence (Buchanan et al., 1992). Affective processes and response to

reward/punishment, which contribute substantially to decision-making, may present

adolescence specific features that facilitate risk-taking behaviour in this age group (Martin

et al., 2002). The cognitive function also develops throughout adolescence. Although this

34

function improves dramatically, qualitatively as well as quantitatively, from infancy into

childhood, it continues to be refined across adolescence (Ernst and Mueller, 2008).

Among the transitional processes seen at some point during adolescence are

puberty and its concomitant hormonal and physiological changes, along with considerable

growth spurt (Spear et al., 2002; Spear, 2007). The initiation of puberty is determined by

an increase, after the prepubertal quiescence period, in the pulsatile pattern of

hypothalamic GnRH secretion, and activation of HPG axis, it culminates in gonadal

maturation and in total capacity to express reproductive behaviour (Payne et al., 1977;

Wiemann et al., 1989; Sisk and Foster, 2004).

It is recognized that in addition to the well-known perinatal period of steroid-

dependent organization of neural circuits and behaviour, adolescence is another period of

development during which steroid hormones organize the nervous system (Sisk and Zehr,

2005). As brain-hormone interactions during the adolescent brain “growth spurt” are

integral to behavioural maturation, temporal dissociations between gonadal maturation

and adolescent brain development are likely to have consequences for adult behaviour

(Sisk and Foster, 2004). In fact, the absence of gonadal hormones or pharmacological

blockade of their action during adolescence prohibits adult-typical expression of behaviour

that are organized during adolescence, even if hormones are replaced in adulthood,

demonstrating irreversible and adverse consequences for behaviour if gonadal hormones

are absent during adolescence (Sisk and Foster, 2004).

Despite the importance of gonadal hormones, it is clear that some important

aspects of behavioural maturation are not driven solely by the appearance of steroid

hormones at the time of puberty (Sisk and Foster, 2004). Structural remodeling of the

brain during adolescence occurs through both steroid-dependent and steroid-independent

mechanisms (Sisk and Foster, 2004).

Both the imaging work in humans and the often more detailed neuroanatomical

assessments emerging from studies with laboratory animals have revealed a surprising

degree of neural sculpting during adolescence, with notable similarities in the brain

regions affected often emerging across a variety of species (Spear, 2007).

During this period there is a massive loss of synapses in neocortical brain regions

(Huttenlocher, 1984; Spear, 2000). Among the synapses undergoing particularly notable

pruning during adolescence are those providing excitatory input to the neocortex

(Bourgeois et al., 1994), as well as synapses contributing to reverberating circuits within

particular cortical regions (Woo et al., 1997). This ontogenetic reduction in excitatory input

35

to cortex and connectivity within reverberating circuitry could contribute to the reduction

and refinement of brain effort during adolescence (Spear, 2007).

Progressive myelination of axons results in considerable developmental increases

in cortical white matter through adolescence and into adulthood (Sowell et al., 2003),

providing faster communication between regions, and has been presumed to increase

overall speed of information processing within the brain. Although less prominent and

consistent than developmental increases in white matter, ontogenetic declines in volume

of gray matter are seen in regions such as the frontal cortex (Giedd et al., 1999; Rapoport

et al., 1999).

Among the brain regions showing particularly marked ontogenetic alterations

during adolescence is the prefrontal cortex, with this region showing considerable synaptic

culling and a prominent loss of excitatory input (Bourgeois et al., 1994). In contrast to the

adolescent-associated loss of excitatory drive to cortex (Zecevic et al., 1989), DA input to

prefrontal cortex increases during adolescence (Kalsbeek et al., 1988; Rosenberg and

Lewis, 1994).

During adolescence certain subcortical regions also undergo considerable

remodeling, especially those regions that form part of an interconnecting network of

circuitry with prefrontal cortex (Spear, 2000). For instance, projections from the amygdala

to prefrontal cortex continue to be elaborated throughout adolescence (Woo et al., 1997;

Cunningham et al., 2002). The amygdala of the adolescent also shows a different pattern

of stress-induced activation compared to the adult (Kellogg et al., 1998).

Among the numerous alterations seen in the mesocorticolimbic brain regions

during adolescence are regionally specific ontogenetic alterations in the patterns of DA

production and utilization. Estimates of DA synthesis and turnover on prefrontal cortex are

higher early in adolescence than later in adolescence and in adulthood, whereas DA

synthesis and/or turnover estimates in Nac and striatum conversely are lower earlier than

late in adolescence (Teicher et al., 1993; Andersen et al., 1997).

The HPA axis, known to be, as already mentionated in this chapter, one of the

main systems regulating physiological responses to stressors, is also in transition during

adolescence. For example, plasma concentrations of corticosterone remains elevated

longer after exposure to a stressor in adolescent rats compared to adults, and sex

steroids regulate HPA axis function in adults but not in adolescents (reviewed in

McCormick et al., 2008). Glucocorticoids affect ongoing behavioural and

neurophysiological processes and can shape future function by altering synaptogenesis,

dendritic morphology and neurogenesis (Joels et al., 2004). Altered HPA axis function in

36

adolescence may make adolescents particularly susceptible to the negative

consequences of stressors.

1.6.1. Adolescence: A critical period of addiction vulnerability

As mentioned above, evidences suggest that human adolescents are more

involved in reckless behaviours than adults and are motivated to seek novel sensations

(Irwin, 1989; La Greca et al., 2001; Martin et al., 2002). Even when considering an animal

model of adolescence in rat, animals undergoing this transition period exhibit more risk

taking, novelty-seeking, and peer-directed social interactions than adults (Douglas et al.,

2003, 2004; Stansfield and Kirstein, 2006). Studies have shown that high levels of

novelty/sensation-seeking are powerful predictors of risky activities such as hazardous

driving, unprotected sex and drug use (Andrucci et al., 1989; Wills et al., 1994). Hence,

along with increased sensation and novelty seeking during adolescence, an increase in

drug use is seen as well (Spear, 2000).

In fact, epidemiological data indicate that experimentation with addictive drugs and

the onset of addictive disorders are primarily concentrated in the adolescence and early

adulthood (Chen and Kandel, 1995; Chambers et al., 2003). Furthermore, adolescents are

more likely to progress from use to addiction state than adults (Chen et al., 1997) and

demonstrate a more precipitous progression of drug use (Estroff et al., 1989; Warner et

al., 1995). There is growing evidence suggesting that adolescence represents a period of

heightened biological vulnerability to the addictive properties of drugs of abuse (Chambers

et al., 2003).

Indeed, adolescents of a variety of species differ in their sensitivity to various drugs

relative to their adult and sometimes younger counterparts (Spear, 2000). Adolescent rats

and mice are less sensitive than younger animals or adult to the effects of psychomotor

stimulants such as amphetamine and cocaine when indexed in terms of acute locomotor

stimulation and stereotypy (Laviola et al., 1995; Bolanos et al., 1998; Laviola et al., 1999)

Adolescent male rats also are more likely to acquire self-administration of nicotine plus

acetaldehyde than adult male rats (Belluzzi et al., 2005). In addition to higher levels of

self-administration compared to adults, adolescent rats also exhibit reduced evidence of

aversive drug effects. The conditioned taste aversion to amphetamine is reduced in

adolescent rats (Infurna and Spear, 1979). Adolescence is also a time of relative

insensitivity to the aversive properties of cocaine (Schramm-Sapyta et al., 2006).

Adolescents require close to 75% more extinction trials than adults to extinguish cocaine

place-preferences compared with adults (Brenhouse and Andersen, 2008). These

37

examples, demonstrate that adolescents are less sensitive to use-limiting effects of drugs,

and therefore may tolerate higher levels than adults which may facilitate development of

addiction (Schramm-Sapyta et al., 2006).

As already mentioned, the brain of the adolescent differs considerably from the

adult in a number of neural systems, namely in systems implicated in the action of drugs

(Spear, 2000). In fact, the mesocorticolimbic dopaminergic brain region, known as taking

part of the circuitry implicated in drug reward processes (Koob, 1992b; Robinson and

Berridge, 2003) is among the neural systems known to undergo developmental alterations

during adolescence (Spear, 2000).

Adolescence is a critical period of transition from a more emotional regulation of

the structures that mediate substance abuse, to a more mature cortical regulatory

mechanism (Spear, 2000). During adolescence, the primary dopaminergic projections to

the Nac extend from the VTA and are predominately modulated by the amygdala (Oades

and Halliday, 1987). However, in adulthood, this previously amygdaloid-modulated system

receives projections from the medial prefrontal cortex, and this developmental transition is

critical in the functional nature of the system (Cunningham et al., 2002).The development

of this system allows for a transition from more emotionally directed behaviour to more

contextually regulated behaviour. Because adolescents lack sufficient cortical regulation,

provided by the medial prefrontal cortex, their behaviour tends to be more impulsive and

guided by emotion than adults, increasing the chances of risky behaviours, including

initiating drug use (Chambers et al., 2003).

Most of what it is known about the consequences of developmental exposure to

drugs and other chemicals is based on exposures during prenatal and early postnatal

period, with little emphasis on exposure periods that include adolescence (Catlow and

Kirstein, 2007; Spear, 2007; Lubman and Yucel, 2008). Given that drug use frequently

occurs during adolescence, and with the increasing recognition that adolescence is a time

of considerable neural restructuring and sculpting of the brain (Spear, 2000), there is,

likewise, a growing interest in assessing whether this developmental transition represents

an unusually sensitive period to disruption by drugs, or whether this remodeling reflects a

window of opportunity for unusual plasticity and recovery (Spear, 2000; Catlow and

Kirstein, 2007; Spear, 2007). Nevertheless, growing evidence from the developmental

neuroscience field suggests that early onset of drug use may adversely affect ongoing

brain development and the critical maturational processes that are occurring during the

adolescent period (Lubman et al., 2007). The following table (table 1.6.1.1) summarizes

the most significant effects of cocaine exposure during adolescence on the rat animal

model.

38

Table 1.6.1.1- Summary of the most common effects of cocaine exposure during adolescence in the rat.

Report Route of administration

Daily dose Exposure period

Evaluation period

Observed effects

(Philpot and Kirstein, 1999)

i.p. 2 or 20 mg/kg/day

PND 21-24

PND 25 (after challenge injection)

↑ followed by suppression in DA overflow in the Nac.

(Collins and Izenwasser, 2002)

i.p. 50 mg/kg/day

PND 28-35

PND45 (after challenge injection)

No sensitization to locomotor activity;

No effects on either DAT or SERT densities.

(Catlow and Kirstein, 2007)

i.p. 20 mg/kg/day

PND 35-44

PND 70-79

PND 65

PND 100

↑DA levels in response to sucrose intake compared to saline and adult.

(Frantz et al., 2007)

i.p.

i.v. catheter

10 or 20 mg/kg; four injections spaced 5 days apart

0.37, 0.74 and 2.92 mg/kg; under a fixed ratio 1 lever-response contingency- 2 hour session

PND 37-52

PND 75-90

PND 37-52

PND75-90

PND 37-52

PND 75-90

PND 37-52

PND 75-90

↓ sensitivity to acute cocaine compared to adult;

↓ overall activity compared to adults;

No effects on cocaine-stimulated Nac DA.

(Marin et al., 2008)

i.p. 10 mg/kg/day

PND 30-34 PND 37, 64 and 97(All preceded by a challenge injection )

Behavioral sensitization on PND37 e 64;

↑ GluR1 in mPFC on PND37 in sensitized rats;

No effects on TH or NR1 in Nac or mPFC.

(Walker and Kuhn, 2008)

ip 15 mg/kg/day

PND 28

PND 65

PND 28

PND 65

↑ DA release in caudate nucleus compared to adult rats.

(Catlow and Kirstein, 2005)

i.p 20 mg/kg/day

PND 35

PND 60

PND 35

PND 60

↑ locomotor activity in the adolescent females compared to adults

(Lepsch et al., 2005)

i.p. 10 mg/kg/day

PND 38 PND 38 Pre-exposure to both chronic restraint and variable stress ↑ cocaine-induced locomotor activity

(Maldonado and Kirstein, 2005)

i.p. 30 mg/kg/day

PND 45

PND 60

PND 45

PND 60

Prior handling ↑ cocaine-induced locomotor activity, but not in adults.

DA- dopamine; DAT- dopamine transporter; GluR1- GluR1 glutamate receptor subunit; i.p.- intraperitoneal injection; i.v.- intravenous injection; mPFC- medial prefrontal cortex; Nac- nucleus accumbens; NR1- NR1 glutamate receptor subunit; PND- postnatal day; SERT- serotonin transporter; TH- tyrosine hydroxilase; ↑ increase; ↓ decrease.

39

Table 1.6.1.1- Summary of the most common effects of cocaine exposure during adolescence in the rat (continuation).

Report Route of administration

Daily dose Exposure period

Evaluation period

Observed effects

(Caster et al., 2007)

i.p. 40 mg/kg/day

PND 28

PND 42

PND 65

PND 28-29

PND 42-43

PND 65-66

Behavioral sensitization to non-ambulatory horizontal activity, sniffing behaviors, and stereotypies in animals of all ages;

Ambulatory sensitization and greater increases in stereotypies observed in the youngest adolescents;

(Badanich et al., 2008)

i.p. 5, 10 or 20 mg/kg/day

PND 35

PND 45

PND 60

PND 35

PND 45

PND 60

↑ acute cocaine-induced locomotor activity in early adolescence comparison to late adolescent and adult rats.

(Badanich et al., 2006)

i.p. 5 or 20 mg/kg/day

PND 31-34

PND 41-44

PND 56-59

PND 35

PND 45

PND 60

Cocaine CPP were found for all ages at the high dose;

PND 35 was the only age group to have a preference at the low dose.

(Schramm-Sapyta et al., 2006)

i.p. 10-40 mg/kg/day

PND 29

PND 66

PND 30

PND 67

↓ susceptibility to cocaine CTA compared to adult.

(Perry et al., 2007)

i.v. catheter

0.4 mg/kg under fixed ration 1 lever-response contingency

PND 29- until acquisition , or PND 59

PND 154- until acquisition or, PND 184

PND 29-59

PND 154-184

↑ avidity for sweets in adolescents;

↑ vulnerability to initiate drug abuse in adolescents.

(Brenhouse and Andersen, 2008)

i.p. 10 mg/kg/day

20 mg/kg/day

PND 39-40

PND 78-79

PND 41 until CPP extinction

PND 80 until CPP extinction

↑ extinction trials than adults to extinguish CPP;

Once extinguished, adolescents display a greater preference for a previously cocaine-paired environment.

CPP-conditioned place preference; CTA- conditioned aversive properties; i.p.- intraperitoneal injection; i.v.- intravenous injection; PND- postnatal day; ↑ increase; ↓ decrease.

40

Table 1.6.1.1- Summary of the most common effects of cocaine exposure during adolescence in the rat (continuation).

Report Route of administration

Daily dose Exposure period

Evaluation period

Observed effects

(Lynch, 2008) i.v. catheter

0.75 mg/kg under a fixed ratio 1 lever-response contingency

PND 30- until acquisition or PND 50

PND 30- until acquisition or PND 50

Females acquired cocaine self-administration more rapid than males;

A greater % of females acquired self-administration;

Response was positively associated with levels of estradiol.

(Zakharova et al., 2009)

i.p. 3, 5, 7.5, 10 or 20 mg/kg/day

PND 35-37

PND 67-69

PND 38

PND 70

Adolescent and adult male rats developed CPP;

The dose-effect curve for cocaine CPP was shifted to the left in adolescents compared to adults.

(Kerstetter and Kantak, 2007)

i.v. catheter

1 mg/kg under a fixed ratio 5 lever-response contingency- 2h sessions

Starting PND 37- 18 sessions

Starting PND 74-79- 18 sessions

18 days later

Normal learning in the conditioned cue preference task in adolescents;

Adults present deficit learning.

(Black et al., 2006)

i.p. 3x5 mg/kg/day from PND35-36;

3x10 mg/kg/day from PND37-39;

2 days of abstinence

3x15 mg/kg/day from PND 42-46;

PND 35-46 PND 56-57

PND 70-71

↑ responsiveness to the stimulant effects of cocaine;

Abnormally rapid shifts in attention in an attentional set-shifting task;

Acute alterations in the expression of genes in the PFC.

(Santucci et al., 2004)

s.c. 10 mg/kg/day

20 mg/kg/day

PND 26-33 PND 44

PND142-150

PND 158-166

PND 410-416

Residual and temporary deleterious effects on memory.

(Santucci, 2008)

s.c. 20 mg/kg/day PND 28-35 About 5 and 9 weeks later

Residual impairs in working memory;

↑ fear memory.

CPP- cocaine place-preferences; i.p.- intraperitoneal; i.v.- intravenous; PFC- prefrontal cortex; PND - postnatal day; s.c.-

subcutaneous injection;; ↑- increase.

41

Table 1.6.1.1- Summary of the most common effects of cocaine exposure during adolescence in the rat (continuation).

Report Route of administration

Daily dose Exposure period

Evaluation period

Observed effects

(Stansfield and Kirstein, 2005)

i.p. 20 mg/kg/day

PND 35

PND 60

PND 35

PND 60

Rapid approach to novel object compared to adult.

(Stansfield and Kirstein, 2007)

i.p. 20 mg/kg/day

PND 30-50 PND 66-69

↑locomotor response in novel environment;

↓time in the center of the novel open field

↓time with a novel object;

Anxiogenic response to novelty.

(Santucci and Madeira, 2008)

s.c. 10 mg/kg/day

PND 30-37 PND 92-97

PND 110-119

Anxiogenic response persists 12 weeks beyond cessation of drug treatment.

(Raap et al., 2000)

s.c. 20 mg/kg/day

PND 21-60 PND 25-80 No effects on onset of puberty;

No effects on date of first ovulation;

↑ number of cycles with no clear proestrus-estrus transition.

i.p. - intraperitoneal; PND - postnatal day; s.c.- subcutaneous; ↑ - increase; ↓ - decrease.

The data presented in the table 1.6.1.1 seems to indicate that adolescence

represents a period of heightened biological vulnerability to the addictive properties of

cocaine, also providing information that cocaine use during adolescence may have

neurobehavioural long-lasting effects.

In addition, it is evident in the overview of the literature references, and as

mentioned by several authors (e.g. Donovan et al., 2001; Catlow and Kirstein, 2007;

Spear, 2007; Lubman and Yucel, 2008) that the characterization of the effects of cocaine

upon the developing adolescent require further investigation in order to the full

understanding of the processes underlying the greater vulnerability to the addictive

properties of drugs during this developmental period.

42

1.7. Objectives

The objective of the present study was to contribute to the further characterization

of the effects of cocaine during adolescence, knows to be a sensitive period in what

concerns to drug problems. To achieve this aim, a series of experimental protocols were

developed in order to characterize short and long-term neurochemical, hormonal and

behavioural effects of chronic cocaine exposure during adolescence in a rat animal model.

The specific objectives of this thesis were:

- To evaluate the short and long-term effects of chronic cocaine exposure during the

adolescence period on the activity of the HPA and the HPG axes of both male and female

wistar rats, by assessing the hormonal plasma level 30 minutes, 1 day, 5 days and 10

days after last administration of cocaine, and by studying, 10 days after last

administration, the expression of genes known to be involved in the activity of the axes;

- To evaluate the short and long-term effects of chronic cocaine exposure during

adolescence period on the dopaminergic and serotonergic systems of both male and

female wistar rats, by assessing the monoamines levels, in areas known to be involved

with drug addiction, 30 minutes, 1 day, 5 days and 10 days after last administration of

cocaine, and by studying, 10 days after last administration, the expression of genes

related with monoaminergic system;

- To evaluate the short and long-term effects of chronic cocaine exposure during

adolescence period, on behavioural strategies adopted to cope with an environmental

anxiogenic situation, by submitting the animals, 2, 5 and 10 days after last administration,

to the elevated plus maze test (EPM), and related the behavioural data with the HPA axis

and monoaminergic system activity, by measuring hormonal and neurochemical

alterations induced by the test;

- To evaluate the presence of depressive-like symptoms during acute withdrawal from

chronic cocaine exposure during adolescence, both in male and female adolescent rats,

by submitting the animals, 2 days after last administration, to the forced-swim test (FST),

and related the behavioural data obtained with the HPA axis and monoaminergic system

activity, by measuring hormonal and neurochemical alterations induced by the test;

- To evaluate the long-term effects of chronic cocaine exposure during adolescence on

the aggressive behaviour of male rats, using a resident-intruder paradigm, and relate the

behavioural data with the HPA axis and monoaminergic system activity, by measuring

hormonal and neurochemical alterations induced by the test;

43

- To determine the relevance of gender-related differences in the responsiveness to the

chronic cocaine exposure during adolescence.

44

1.8. References

Abel EL, Moore C, Waselewsky D, Zajac C, Russell LD (1989) Effects of cocaine hydrochloride on reproductive function and sexual behavior of male rats and on the behavior of their offspring. J Androl 10:17-27.

Ahima RS, Lawson AN, Osei SY, Harlan RE (1992) Sexual dimorphism in regulation of type II corticosteroid receptor immunoreactivity in the rat hippocampus. Endocrinology 131:1409-1416.

Akana SF, Dallman MF, Bradbury MJ, Scribner KA, Strack AM, Walker CD (1992a) Feedback and facilitation in the adrenocortical system: unmasking facilitation by partial inhibition of the glucocorticoid response to prior stress. Endocrinology 131:57-68.

Akana SF, Scribner KA, Bradbury MJ, Strack AM, Walker CD, Dallman MF (1992b) Feedback sensitivity of the rat hypothalamo-pituitary-adrenal axis and its capacity to adjust to exogenous corticosterone. Endocrinology 131:585-594.

Alexander SL, Irvine CH, Donald RA (1996) Dynamics of the regulation of the hypothalamo-pituitary-adrenal (HPA) axis determined using a nonsurgical method for collecting pituitary venous blood from horses. Front Neuroendocrinol 17:1-50.

Andersen SL, Teicher MH (2000) Sex differences in dopamine receptors and their relevance to ADHD. Neurosci Biobehav Rev 24:137-141.

Andersen SL, Dumont NL, Teicher MH (1997) Developmental differences in dopamine synthesis inhibition by (+/-)-7-OH-DPAT. Naunyn Schmiedebergs Arch Pharmacol 356:173-181.

Andrucci GL, Archer RP, Pancoast DL, Gordon RA (1989) The relationship of MMPI and sensation seeking scales to adolescent drug use. J Pers Assess 53:253-266.

Antoni FA (1993) Vasopressinergic control of pituitary adrenocorticotropin secretion comes of age. Front Neuroendocrinol 14:76-122.

Atkinson HC, Waddell BJ (1997) Circadian variation in basal plasma corticosterone and adrenocorticotropin in the rat: sexual dimorphism and changes across the estrous cycle. Endocrinology 138:3842-3848.

Attardi B, Tsujii T, Friedman R, Zeng Z, Roberts JL, Dellovade T, Pfaff DW, Chandran UR, Sullivan MW, DeFranco DB (1997) Glucocorticoid repression of gonadotropin-releasing hormone gene expression and secretion in morphologically distinct subpopulations of GT1-7 cells. Mol Cell Endocrinol 131:241-255.

Badanich KA, Adler KJ, Kirstein CL (2006) Adolescents differ from adults in cocaine conditioned place preference and cocaine-induced dopamine in the nucleus accumbens septi. Eur J Pharmacol 550:95-106.

Badanich KA, Maldonado AM, Kirstein CL (2008) Early adolescents show enhanced acute cocaine-induced locomotor activity in comparison to late adolescent and adult rats. Dev Psychobiol 50:127-133.

Bagatell CJ, Heiman JR, Rivier JE, Bremner WJ (1994) Effects of endogenous testosterone and estradiol on sexual behavior in normal young men. J Clin Endocrinol Metab 78:711-716.

Balopole DC, Hansult CD, Dorph D (1979) Effect of cocaine on food intake in rats. Psychopharmacology (Berl) 64:121-122.

Barbacka-Surowiak G, Surowiak J, Stoklosowa S (2003) The involvement of suprachiasmatic nuclei in the regulation of estrous cycles in rodents. Reprod Biol 3:99-129.

Barnes NM, Sharp T (1999) A review of central 5-HT receptors and their function. Neuropharmacology 38:1083-1152.

Baumann M, Becketts K, Rothman R (1995a) Evidence for alterations in presynaptic serotonergic function during withdrawal from chronic cocaine in rats. Eur J Pharmacol 282:87-93.

Baumann M, Gendron T, Becketts K, Henningfield J, Gorelick D, Rothman R (1995b) Effects of intravenous cocaine on plasma cortisol and prolactin in human cocaine abusers. Biol Psychiatry 38:751-755.

45

Becker JB (1990a) Direct effect of 17 beta-estradiol on striatum: sex differences in dopamine release. Synapse 5:157-164.

Becker JB (1990b) Estrogen rapidly potentiates amphetamine-induced striatal dopamine release and rotational behavior during microdialysis. Neurosci Lett 118:169-171.

Becker JB (1999) Gender differences in dopaminergic function in striatum and nucleus accumbens. Pharmacol Biochem Behav 64:803-812.

Becker JB, Ramirez VD (1981) Sex differences in the amphetamine stimulated release of catecholamines from rat striatal tissue in vitro. Brain Res 204:361-372.

Beiko J, Lander R, Hampson E, Boon F, Cain DP (2004) Contribution of sex differences in the acute stress response to sex differences in water maze performance in the rat. Behav Brain Res 151:239-253.

Belluzzi JD, Wang R, Leslie FM (2005) Acetaldehyde enhances acquisition of nicotine self-administration in adolescent rats. Neuropsychopharmacology 30:705-712.

Bethea CL, Pecins-Thompson M, Schutzer WE, Gundlah C, Lu ZN (1998) Ovarian steroids and serotonin neural function. Mol Neurobiol 18:87-123.

Bhattacharyya AK, Pradhan SN (1979) Interactions between motor activity and sterotypy in cocaine-treated rats. Psychopharmacology (Berl) 63:311-312.

Bingaman EW, Magnuson DJ, Gray TS, Handa RJ (1994) Androgen inhibits the increases in hypothalamic corticotropin-releasing hormone (CRH) and CRH-immunoreactivity following gonadectomy. Neuroendocrinology 59:228-234.

Birmingham MK, Sar M, Stumpf WE (1984) Localization of aldosterone and corticosterone in the central nervous system, assessed by quantitative autoradiography. Neurochem Res 9:333-350.

Black YD, Maclaren FR, Naydenov AV, Carlezon WA, Jr., Baxter MG, Konradi C (2006) Altered attention and prefrontal cortex gene expression in rats after binge-like exposure to cocaine during adolescence. J Neurosci 26:9656-9665.

Bolanos C, Glatt S, Jackson D (1998) Subsensitivity to dopaminergic drugs in periadolescent rats: a behavioral and neurochemical analysis. Brain Res Dev Brain Res 111:25-33.

Booze RM, Wood ML, Welch MA, Berry S, Mactutus CF (1999) Estrous cyclicity and behavioral sensitization in female rats following repeated intravenous cocaine administration. Pharmacol Biochem Behav 64:605-610.

Borowsky B, Kuhn CM (1991a) Chronic cocaine administration sensitizes behavioral but not neuroendocrine responses. Brain Res 543:301-306.

Borowsky B, Kuhn CM (1991b) Monoamine mediation of cocaine-induced hypothalamo-pituitary-adrenal activation. J Pharmacol Exp Ther 256:204-210.

Bourgeois JP, Goldman-Rakic PS, Rakic P (1994) Synaptogenesis in the prefrontal cortex of rhesus monkeys. Cereb Cortex 4:78-96.

Bowman BP, Vaughan SR, Walker QD, Davis SL, Little PJ, Scheffler NM, Thomas BF, Kuhn CM (1999) Effects of sex and gonadectomy on cocaine metabolism in the rat. J Pharmacol Exp Ther 290:1316-1298.

Breen KM, Stackpole CA, Clarke IJ, Pytiak AV, Tilbrook AJ, Wagenmaker ER, Young EA, Karsch FJ (2004) Does the type II glucocorticoid receptor mediate cortisol-induced suppression in pituitary responsiveness to gonadotropin-releasing hormone? Endocrinology 145:2739-2746.

Brenhouse HC, Andersen SL (2008) Delayed extinction and stronger reinstatement of cocaine conditioned place preference in adolescent rats, compared to adults. Behav Neurosci 122:460-465.

Briski KP, Sylvester PW (1991) Acute inhibition of pituitary LH release in the male rat by the glucocorticoid agonist decadron phosphate. Neuroendocrinology 54:313-320.

46

Broadbear JH, Winger G, Cicero TJ, Woods JH (1999a) Effects of self-administered cocaine on plasma adrenocorticotropic hormone and cortisol in male rhesus monkeys. J Pharmacol Exp Ther 289:1641-1647.

Broadbear JH, Winger G, Cicero TJ, Woods JH (1999b) Effects of response contingent and noncontingent cocaine injection on hypothalamic-pituitary-adrenal activity in rhesus monkeys. J Pharmacol Exp Ther 290:393-402.

Broderick PA, Rahni DN, Zhou Y (2003) Acute and subacute effects of risperidone and cocaine on accumbens dopamine and serotonin release using in vivo microvoltammetry on line with open-field behavior. Prog Neuropsychopharmacol Biol Psychiatry 27:1037-1054.

Buchanan CM, Eccles JS, Becker JB (1992) Are adolescents the victims of raging hormones: evidence for activational effects of hormones on moods and behavior at adolescence. Psychol Bull 111:62-107.

Burgess LH, Handa RJ (1992) Chronic estrogen-induced alterations in adrenocorticotropin and corticosterone secretion, and glucocorticoid receptor-mediated functions in female rats. Endocrinology 131:1261-1269.

Buydens-Branchey L, Branchey M, Fergeson P, Hudson J, McKernin C (1997) Craving for cocaine in addicted users. Role of serotonergic mechanisms. Am J Addict 6:65-73.

Buydens-Branchey L, Branchey M, Hudson J, Rothman M, Fergeson P, McKernin C (1998) Effect of fenfluramine challenge on cocaine craving in addicted male users. Am J Addict 7:142-155.

Cabrera R, Diaz A, Pinter A, Belmar J (1993) In vitro progesterone effects on 3H-dopamine release from rat corpus striatum slices obtained under different endocrine conditions. Life Sci 53:1767-1777.

Caine SB, Koob GF (1994) Effects of dopamine D-1 and D-2 antagonists on cocaine self-administration under different schedules of reinforcement in the rat. J Pharmacol Exp Ther 270:209-218.

Callahan P, Cunningham K (1995) Modulation of the discriminative stimulus properties of cocaine by 5- HT1B and 5-HT2C receptors. J Pharmacol Exp Ther 274:1414-1424.

Calogero AE, Gallucci WT, Kling MA, Chrousos GP, Gold PW (1989) Cocaine stimulates rat hypothalamic corticotropin-releasing hormone secretion in vitro. Brain Res 505:7-11.

Calogero AE, Burrello N, Bosboom AM, Garofalo MR, Weber RF, D'Agata R (1999) Glucocorticoids inhibit gonadotropin-releasing hormone by acting directly at the hypothalamic level. J Endocrinol Invest 22:666-670.

Carboni E, Imperato A, Perezzani L, Di Chiara G (1989) Amphetamine, cocaine, phencyclidine and nomifensine increase extracellular dopamine concentrations preferentially in the nucleus accumbens of freely moving rats. Neuroscience 28:653-661.

Carey MP, Deterd CH, de Koning J, Helmerhorst F, de Kloet ER (1995) The influence of ovarian steroids on hypothalamic-pituitary-adrenal regulation in the female rat. J Endocrinol 144:311-321.

Carey R, DePalma G, Damianopoulos E (2001) Cocaine and serotonin: a role for the 5-HT(1A) receptor site in the mediation of cocaine stimulant effects. Behav Brain Res 126:127-133.

Carr KD, Kim GY, Cabeza de Vaca S (2000) Chronic food restriction in rats augments the central rewarding effect of cocaine and the delta1 opioid agonist, DPDPE, but not the delta2 agonist, deltorphin-II. Psychopharmacology (Berl) 152:200-207.

Carroll ME, Lac ST, Asencio M, Kragh R (1990) Intravenous cocaine self-administration in rats is reduced by dietary L-tryptophan. Psychopharmacology (Berl) 100:293-300.

Cass WA, Gerhardt GA, Mayfield RD, Curella P, Zahniser NR (1992) Differences in dopamine clearance and diffusion in rat striatum and nucleus accumbens following systemic cocaine administration. J Neurochem 59:259-266.

47

Caster JM, Walker QD, Kuhn CM (2007) A single high dose of cocaine induces differential sensitization to specific behaviors across adolescence. Psychopharmacology (Berl) 193:247-260.

Catlow BJ, Kirstein CL (2005) Heightened cocaine-induced locomotor activity in adolescent compared to adult female rats. J Psychopharmacol 19:443-447.

Catlow BJ, Kirstein CL (2007) Cocaine during adolescence enhances dopamine in response to a natural reinforcer. Neurotoxicol Teratol 29:57-65.

Ceccatelli S, Cintra A, Hokfelt T, Fuxe K, Wikstrom AC, Gustafsson JA (1989) Coexistence of glucocorticoid receptor-like immunoreactivity with neuropeptides in the hypothalamic paraventricular nucleus. Exp Brain Res 78:33-42.

Chambers RA, Taylor JR, Potenza MN (2003) Developmental neurocircuitry of motivation in adolescence: A critical period of addiction vulnerability. Am J Psychiatry 160:1041-1052.

Chappell PE, Lee J, Levine JE (2000) Stimulation of gonadotropin-releasing hormone surges by estrogen. II. Role of cyclic adenosine 3'5'-monophosphate. Endocrinology 141:1486-1492.

Chen JP, van Praag HM, Gardner EL (1991) Activation of 5-HT3 receptor by 1-phenylbiguanide increases dopamine release in the rat nucleus accumbens. Brain Res 543:354-357.

Chen K, Kandel DB (1995) The natural history of drug use from adolescence to the mid-thirties in a general population sample. Am J Public Health 85:41-47.

Chen K, Kandel D (2002) Relationship between extent of cocaine use and dependence among adolescents and adults in the United States. Drug Alcohol Depend 68:65-85.

Chen K, Kandel DB, Davies M (1997) Relationships between frequency and quantity of marijuana use and last year proxy dependence among adolescents and adults in the United States. Drug Alcohol Depend 46:53-67.

Chen R, Osterhaus G, McKerchar T, Fowler SC (2003) The role of exogenous testosterone in cocaine-induced behavioral sensitization and plasmalemmal or vesicular dopamine uptake in castrated rats. Neuroscience Letters 351:161-164.

Chen X, Herbert J (1995) The effect of long-term castration on the neuronal and physiological responses to acute or repeated restraint stress: interactions with opioids and prostaglandins. J Neuroendocrinol 7:137-144.

Chin J, Sternin O, Wu HB, Fletcher H, Perrotti LI, Jenab S, Quinones-Jenab V (2001) Sex differences in cocaine-induced behavioral sensitization. Cell Mol Biol 47:1089-1095.

Chin J, Sternin O, Wu H, Burrell S, Lu D, Jenab S, Perrotti L, Quinones-Jenab V (2002) Endogenous gonadal hormones modulate behavioral and neurochemical responses to acute and chronic cocaine administration. Brain Res 945:123-130.

Chrousos GP, Gold PW (1998) A healthy body in a healthy mind--and vice versa--the damaging power of "uncontrollable" stress. J Clin Endocrinol Metab 83:1842-1845.

Church WH, Justice JB, Jr., Byrd LD (1987) Extracellular dopamine in rat striatum following uptake inhibition by cocaine, nomifensine and benztropine. Eur J Pharmacol 139:345-348.

Cintra A, Zoli M, Rosen L, Agnati LF, Okret S, Wikstrom AC, Gustaffsson JA, Fuxe K (1994) Mapping and computer assisted morphometry and microdensitometry of glucocorticoid receptor immunoreactive neurons and glial cells in the rat central nervous system. Neuroscience 62:843-897.

Cintra A, Fuxe K, Solfrini V, Agnati LF, Tinner B, Wikstrom AC, Staines W, Okret S, Gustafsson JA (1991) Central peptidergic neurons as targets for glucocorticoid action. Evidence for the presence of glucocorticoid receptor immunoreactivity in various types of classes of peptidergic neurons. J Steroid Biochem Mol Biol 40:93-103.

Cole MA, Kim PJ, Kalman BA, Spencer RL (2000) Dexamethasone suppression of corticosteroid secretion: evaluation of the site of action by receptor measures and functional studies. Psychoneuroendocrinology 25:151-167.

48

Collins S, Izenwasser S (2002) Cocaine differentially alters behavior and neurochemistry in periadolescent versus adult rats. Brain Res Dev Brain Res 138:27-34.

Costall B, Kelly ME, Naylor RJ, Onaivi ES (1989) The actions of nicotine and cocaine in a mouse model of anxiety. Pharmacol Biochem Behav 33:197-203.

Creese I, Iversen SD (1974) The role of forebrain dopamine systems in amphetamine induced stereotyped behavior in the rat. Psychopharmacologia 39:345-357.

Cunningham KA, Paris JM, Goeders NE (1992) Chronic cocaine enhances serotonin autoregulation and serotonin uptake binding. Synapse 11:112-123.

Cunningham MG, Bhattacharyya S, Benes FM (2002) Amygdalo-cortical sprouting continues into early adulthood: implications for the development of normal and abnormal function during adolescence. J Comp Neurol 453:116-130.

Dackis CA, O'Brien CP (2001) Cocaine dependence: a disease of the brain's reward centers. Journal of Substance Abuse Treatment 21:111-117.

Dallman MF, Akana SF, Cascio CS, Darlington DN, Jacobson L, Levin N (1987) Regulation of ACTH secretion: variations on a theme of B. Recent Prog Horm Res 43:113-173.

Dallman MF, Viau VG, Bhatnagar S, Gomez F, Laugero K, Bell ME (2002) Corticotropin-Releasing Factor, Corticosteroids, Stress, and Sugar: Energy Balance, the Brain, and Behavior. In: Hormones, Brain and Behavior (Pfaff DW, Arnold AP, Etgen AM, Fahrbach SE, Rubin RT, eds). San Diego, California: Academic Press.

Davis CF, Davis BF, Halaris AE (1977) Variations in the uptake of 3H-dopamine during the estrous cycle. Life Sci 20:1319-1332.

De La Garza R, 2nd, Cunningham KA (2000) The effects of the 5-hydroxytryptamine(1A) agonist 8-hydroxy-2-(di-n-propylamino)tetralin on spontaneous activity, cocaine-induced hyperactivity and behavioral sensitization: a microanalysis of locomotor activity. J Pharmacol Exp Ther 292:610-617.

De Wit H, Wise RA (1977) Blockade of cocaine reinforcement in rats with the dopamine receptor blocker pimozide, but not with the noradrenergic blockers phentolamine or phenoxybenzamine. Can J Psychol 31:195-203.

Deroche V, Marinelli M, Le Moal M, Piazza PV (1997) Glucocorticoids and behavioral effects of psychostimulants. II: cocaine intravenous self-administration and reinstatement depend on glucocorticoid levels. J Pharmacol Exp Ther 281:1401-1407.

Dierschke DJ, Hutz RJ, Wolf RC (1985) Induced follicular atresia in rhesus monkeys: strength-duration relationships of the estrogen stimulus. Endocrinology 117:1397-1403.

Diorio D, Viau V, Meaney MJ (1993) The role of the medial prefrontal cortex (cingulate gyrus) in the regulation of hypothalamic-pituitary-adrenal responses to stress. J Neurosci 13:3839-3847.

Donovan B, Padin-Rivera E, Kowaliw S (2001) "Transcend": initial outcomes from a posttraumatic stress disorder/substance abuse treatment program. J Trauma Stress 14:757-772.

Douglas LA, Varlinskaya EI, Spear LP (2003) Novel-object place conditioning in adolescent and adult male and female rats: effects of social isolation. Physiol Behav 80:317-325.

Douglas LA, Varlinskaya EI, Spear LP (2004) Rewarding properties of social interactions in adolescent and adult male and female rats: impact of social versus isolate housing of subjects and partners. Dev Psychobiol 45:153-162.

Elman I, Karlsgodt KH, Gastfriend DR (2001) Gender differences in cocaine craving among non-treatment-seeking individuals with cocaine dependence. Am J Drug Alcohol Abuse 27:193-202.

Elman I, Breiter HC, Gollub RL, Krause S, Kantor HL, Baumgartner WA, Gastfriend DR, Rosen BR (1999) Depressive symptomatology and cocaine-induced pituitary-adrenal axis activation in individuals with cocaine dependence. Drug Alcohol Depend 56:39-45.

EMCDDA (2008) The state of the drugs problems in Europe. Annual report 2008. In, pp 58-67: European Monitoring Centre for Drugs and Drug Addiction

49

Erb S, Shaham Y, Stewart J (1998) The role of corticotropin-releasing factor and corticosterone in stress- and cocaine-induced relapse to cocaine seeking in rats. J Neurosci 18:5529-5536.

Ernst M, Mueller SC (2008) The adolescent brain: insights from functional neuroimaging research. Dev Neurobiol 68:729-743.

Estroff TW, Schwartz RH, Hoffmann NG (1989) Adolescent cocaine abuse- addictive potential, behavioral and psychiatric effects. Clinical Pediatrics 28:550-555.

Evans S, Haney M, Foltin R (2002) The effects of smoked cocaine during the follicular and luteal phases of the menstrual cycle in women. Psychopharmacology (Berl) 159:397-406.

Evans SM, Foltin RW (2006) Pharmacokinetics of repeated doses of intravenous cocaine across the menstrual cycle in rhesus monkeys. Pharmacol Biochem Behav 83:56-66.

Everitt BJ, Wolf ME (2002) Psychomotor stimulant addiction: a neural systems perspective. J Neurosci 22:3312-3320.

Ferris RM, Tang FL, Maxwell RA (1972) A comparison of the capacities of isomers of amphetamine, deoxypipradrol and methylphenidate to inhibit the uptake of tritiated catecholamines into rat cerebral cortex slices, synaptosomal preparations of rat cerebral cortex, hypothalamus and striatum and into adrenergic nerves of rabbit aorta. J Pharmacol Exp Ther 181:407-416.

Festa E, Quinones-Jenab V (2004) Gonadal hormones provide the biological basis for sex differences in behavioral responses to cocaine. Horm Behav 46:509-519.

Festa E, Russo S, Gazi F, Niyomchai T, Kemen L, Lin S, Foltz R, Jenab S, Quinones-Jenab V (2004) Sex differences in cocaine-induced behavioral responses, pharmacokinetics, and monoamine levels. Neuropharmacology 46:672-687.

Festa ED, Jenab S, Chin J, Gazi FM, Wu HBK, Russo SJ, Quinones-Jenab V (2003) Frequency of cocaine administration affects behavioral and endocrine responses in male and female Fischer rats. Cellular and Molecular Biology 49:1275-1280.

Finkelstein JS, Whitcomb RW, O'Dea LS, Longcope C, Schoenfeld DA, Crowley WF, Jr. (1991) Sex steroid control of gonadotropin secretion in the human male. I. Effects of testosterone administration in normal and gonadotropin-releasing hormone-deficient men. J Clin Endocrinol Metab 73:609-620.

Fischman MW, Schuster CR, Rajfer S (1983) A comparison of the subjective and cardiovascular effects of cocaine and procaine in humans. Pharmacol Biochem Behav 18:711-716.

Flugge G, Pfender D, Rudolph S, Jarry H, Fuchs E (1999) 5HT1A-receptor binding in the brain of cyclic and ovariectomized female rats. J Neuroendocrinol 11:243-249.

Fontana DJ, Commissaris RL (1989) Effects of cocaine on conflict behavior in the rat. Life Sci 45:819-827.

Frantz KJ, O'Dell LE, Parsons LH (2007) Behavioral and neurochemical responses to cocaine in periadolescent and adult rats. Neuropsychopharmacology 32:625-637.

Fuxe K, Wikstrom AC, Okret S, Agnati LF, Harfstrand A, Yu ZY, Granholm L, Zoli M, Vale W, Gustafsson JA (1985) Mapping of glucocorticoid receptor immunoreactive neurons in the rat tel- and diencephalon using a monoclonal antibody against rat liver glucocorticoid receptor. Endocrinology 117:1803-1812.

Galici R, Pechnick RN, Poland RE, France CP (2000) Comparison of noncontingent versus contingent cocaine administration on plasma corticosterone levels in rats. Eur J Pharmacol 387:59-62.

Gardi J, Biro E, Sarnyai Z, Vecsernyes M, Julesz J, Telegdy G (1997) Time-dependent alterations in corticotropin-releasing factor-like immunoreactivity in different brain regions after acute cocaine administration to rats. Neuropeptides 31:15-18.

Gawin FH, Kleber HD (1986) Abstinence symptomatology and psychiatric diagnosis in cocaine abusers. Clinical observations. Arch Gen Psychiatry 43:107-113.

Gawin FH, Ellinwood EH, Jr. (1989) Cocaine dependence. Annu Rev Med 40:149-161.

50

Giedd JN, Blumenthal J, Jeffries NO, Castellanos FX, Liu H, Zijdenbos A, Paus T, Evans AC, Rapoport JL (1999) Brain development during childhood and adolescence: a longitudinal MRI study. Nat Neurosci 2:861-863.

Gillies GE, Linton EA, Lowry PJ (1982) Corticotropin releasing activity of the new CRF is potentiated several times by vasopressin. Nature 299:355-357.

Glatz AC, Ehrlich M, Bae RS, Clarke MJ, Quinlan PA, Brown EC, Rada P, Hoebel BG (2002) Inhibition of cocaine self-administration by fluoxetine or D-fenfluramine combined with phentermine. Pharmacol Biochem Behav 71:197-204.

Glick SD, Hinds PA, Shapiro RM (1983) Cocaine-induced rotation: sex-dependent differences between left- and right-sided rats. Science 221:775-777.

Glowinski J, Axelrod J (1965) Effect of drugs on the uptake, release, and metabolism of H3-Norepinephrine in the rat brain. J Pharmacol Exp Ther 149:43-49.

Goeders (2002a) The HPA axis and cocaine reinforcement. Psychoneuroendocrinology 27:13-33.

Goeders N (1997) A neuroendocrine role in cocaine reinforcement. Psychoneuroendocrinology 22:237-259.

Goeders N, Guerin G (1996a) Effects of surgical and pharmacological adrenalectomy on the initiation and maintenance of intravenous cocaine self-administration in rats. Brain Res 722:145-152.

Goeders NE (2002b) Stress and cocaine addiction. J Pharmacol Exp Ther 301:785-789.

Goeders NE, Guerin GF (1996b) Role of corticosterone in intravenous cocaine self-administration in rats. Neuroendocrinology 64:337-348.

Goeders NE, Guerin GF (2000) Effects of the CRH receptor antagonist CP-154,526 on intravenous cocaine self-administration in rats. Neuropsychopharmacology 23:577-586.

Goeders NE, Clampitt DM (2002) Potential role for the hypothalamo-pituitary-adrenal axis in the conditioned reinforcer-induced reinstatement of extinguished cocaine seeking in rats. Psychopharmacology (Berl) 161:222-232.

Goeders NE, Bienvenu OJ, De Souza EB (1990) Chronic cocaine administration alters corticotropin-releasing factor receptors in the rat brain. Brain Res 531:322-328.

Goeders NE, McNulty MA, Guerin GF (1993) Effects of alprazolam on intravenous cocaine self-administration in rats. Pharmacology Biochemistry and Behavior 44:471-474.

Gold PW, Chrousos G, Kellner C, Post R, Roy A, Augerinos P, Schulte H, Oldfield E, Loriaux DL (1984) Psychiatric implications of basic and clinical studies with corticotropin-releasing factor. Am J Psychiatry 141:619-627.

Goodman AL, Hodgen GD (1983) The ovarian triad of the primate menstrual cycle. Recent Prog Horm Res 39:1-73.

Gore AC, Attardi B, DeFranco DB (2006) Glucocorticoid repression of the reproductive axis: effects on GnRH and gonadotropin subunit mRNA levels. Mol Cell Endocrinol 256:40-48.

Guyton AC, Hall JE (2006) Textbook of Medical Physiology, 11th Edition. Philadelphia, Pennsylvania: Elsevier Inc.

Handa RJ, Nunley KM, Lorens SA, Louie JP, McGivern RF, Bollnow MR (1994) Androgen regulation of adrenocorticotropin and corticosterone secretion in the male rat following novelty and foot shock stressors. Physiol Behav 55:117-124.

Haney M, Maccari S, Le Moal M, Simon H, Piazza PV (1995) Social stress increases the acquisition of cocaine self-administration in male and female rats. Brain Res 698:46-52.

Harfstrand A, Fuxe K, Cintra A, Agnati LF, Zini I, Wikstrom AC, Okret S, Yu ZY, Goldstein M, Steinbusch H, et al. (1986) Glucocorticoid receptor immunoreactivity in monoaminergic neurons of rat brain. Proc Natl Acad Sci U S A 83:9779-9783.

51

Harris GC, Altomare K, Aston-Jones G (2001) Preference for a cocaine-associated environment is attenuated by augmented accumbal serotonin in cocaine withdrawn rats. Psychopharmacology (Berl) 156:14-22.

Heesch CM, Negus BH, Keffer JH, Snyder RW, 2nd, Risser RC, Eichhorn EJ (1995) Effects of cocaine on cortisol secretion in humans. Am J Med Sci 310:61-64.

Heesch CM, Negus BH, Bost JE, Keffer JH, Snyder RW, Eichhorn EJ (1996) Effects of cocaine on anterior pituitary and gonadal hormones. J Pharmacol Exp Ther 278:1195-1200.

Heinrichs SC, Menzaghi F, Merlo Pich E, Britton KT, Koob GF (1995) The role of CRF in behavioral aspects of stress. Ann N Y Acad Sci 771:92-104.

Hiroshige T, Abe K, Wada S, Kaneko M (1973) Sex difference in circadian periodicity of CRF activity in the rat hypothalamus. Neuroendocrinology 11:306-320.

Hoplight BJ, Vincow ES, Neumaier JF (2007) Cocaine increases 5-HT1B mRNA in rat nucleus accumbens shell neurons. Neuropharmacology 52:444-449.

Hoyer D, Hannon JP, Martin GR (2002) Molecular, pharmacological and functional diversity of 5-HT receptors. Pharmacol Biochem Behav 71:533-554.

Huttenlocher PR (1984) Synapse elimination and plasticity in developing human cerebral cortex. Am J Ment Defic 88:488-496.

Hyman SE (1996) Addiction to cocaine and amphetamine. Neuron 16:901-904.

Hyman SE, Malenka RC (2001) Addiction and the brain: the neurobiology of compulsion and its persistence. Nat Rev Neurosci 2:695-703.

Imperato A, Puglisi-Allegra S, Casolini P, Zocchi A, Angelucci L (1989) Stress-induced enhancement of dopamine and acetylcholine release in limbic structures: role of corticosterone. Eur J Pharmacol 165:337-338.

Infurna RN, Spear LP (1979) Developmental changes in amphetamine-induced taste aversions. Pharmacol Biochem Behav 11:31-35.

Irwin CE, Jr. (1989) Risk taking behaviors in the adolescent patient: are they impulsive? Pediatr Ann 18:122, 124, 125 passim.

Iversen LL (1973) Catecholamine uptake processes. Br Med Bull 29:130-135.

Jacobsen L, Giedd J, Kreek M, Gottschalk C, Kosten T (2001a) Quantitative medial temporal lobe brain morphology and hypothalamic-pituitary-adrenal axis function in cocaine dependence: a preliminary report. Drug Alcohol Depend 62:49-56.

Jacobsen LK, Southwick SM, Kosten TR (2001b) Substance use disorders in patients with posttraumatic stress disorder: a review of the literature. Am J Psychiatry 158:1184-1190.

Jacobson L, Sapolsky R (1991) The role of the hippocampus in feedback regulation of the hypothalamic-pituitary-adrenocortical axis. Endocr Rev 12:118-134.

Jasper MS, Engeland WC (1994) Splanchnic neural activity modulates ultradian and circadian rhythms in adrenocortical secretion in awake rats. Neuroendocrinology 59:97-109.

Jiang LH, Ashby CR, Jr., Kasser RJ, Wang RY (1990) The effect of intraventricular administration of the 5-HT3 receptor agonist 2-methylserotonin on the release of dopamine in the nucleus accumbens: an in vivo chronocoulometric study. Brain Res 513:156-160.

Joels M, de Kloet ER (1994) Mineralocorticoid and glucocorticoid receptors in the brain. Implications for ion permeability and transmitter systems. Prog Neurobiol 43:1-36.

Joels M, Karst H, Alfarez D, Heine VM, Qin Y, van Riel E, Verkuyl M, Lucassen PJ, Krugers HJ (2004) Effects of chronic stress on structure and cell function in rat hippocampus and hypothalamus. Stress 7:221-231.

Judd SJ, Rigg LA, Yen SS (1979) The effects of ovariectomy and estrogen treatment on the dopamine inhibition of gonadotropin and prolactin release. J Clin Endocrinol Metab 49:182-184.

52

Kalivas P, Duffy P (1993) Time course of extracellular dopamine and behavioral sensitization to cocaine. 1. Dopamine axon terminals. Journal of Neuroscience 13:266-275.

Kalsbeek A, Voorn P, Buijs RM, Pool CW, Uylings HB (1988) Development of the dopaminergic innervation in the prefrontal cortex of the rat. J Comp Neurol 269:58-72.

Kandel D, Chen K, Warner LA, Kessler RC, Grant B (1997) Prevalence and demographic correlates of symptoms of last year dependence on alcohol, nicotine, marijuana and cocaine in the U.S. population. Drug Alcohol Depend 44:11-29.

Kankaanpaa A, Meririnne E, Seppala T (2002) 5-HT3 receptor antagonist MDL 72222 attenuates cocaine- and mazindol-, but not methylphenidate-induced neurochemical and behavioral effects in the rat. Psychopharmacology (Berl) 159:341-350.

Kankaanpaa A, Lillsunde P, Ruotsalainen M, Ahtee L, Seppala T (1996) 5-HT3 receptor antagonist MDL 72222 dose-dependently attenuates cocaine- and amphetamine-induced elevations of extracellular dopamine in the nucleus accumbens and the dorsal striatum. Pharmacol Toxicol 78:317-321.

Karch SB (1996) The pathology of drug abuse. In, pp 1-10. Boca Raton: CRC Press, INC.

Keller-Wood ME, Dallman MF (1984) Corticosteroid inhibition of ACTH secretion. Endocr Rev 5:1-24.

Kellogg CK, Awatramani GB, Piekut DT (1998) Adolescent development alters stressor-induced Fos immunoreactivity in rat brain. Neuroscience 83:681-689.

Kerstetter KA, Kantak KM (2007) Differential effects of self-administered cocaine in adolescent and adult rats on stimulus-reward learning. Psychopharmacology (Berl) 194:403-411.

King GR, Xiong Z, Douglass S, Ellinwood EH (2000) Long-term blockade of the expression of cocaine sensitization by ondansetron, a 5-HT(3) receptor antagonist. Eur J Pharmacol 394:97-101.

King T, Schenken R, Kang I, Javors M, Riehl R (1990) Cocaine disrupts estrous cyclicity and alters the reproductive neuroendocrine axis in the rat. Neuroendocrinology 51:15-22.

King T, McNichol M, Canez M, Javors M, Schenken R (2001) Effect of acute administration of cocaine on pituitary gonadotrophin secretion in female rats. Reproduction 122:723-729.

King TS, Canez MS, Gaskill S, Javors MA, Schenken RS (1993) Chronic cocaine disruption of estrous cyclicity in the rat: dose-dependent effects. J Pharmacol Exp Ther 264:29-34.

Kippin TE, Szumlinski KK, Kapasova Z, Rezner B, See RE (2008) Prenatal stress enhances responsiveness to cocaine. Neuropsychopharmacology 33:769-782.

Kononen J, Honkaniemi J, Gustafsson JA, Pelto-Huikko M (1993) Glucocorticoid receptor colocalization with pituitary hormones in the rat pituitary gland. Mol Cell Endocrinol 93:97-103.

Koob G (1992a) Drugs of abuse: anatomy, pharmacology and function of reward pathways. Trends Pharmacol Sci 13:177-184.

Koob G, Caine S (1999) Cocaine addiction therapy--are we partially there? Nat Med 5:993-995.

Koob GF (1992b) Neural mechanisms of drug reinforcement. Ann N Y Acad Sci 654:171-191.

Koob GF (1999) Stress, Corticotropin-releasing Factor, and Drug Addiction. Ann NY Acad Sci 897:27-45.

Koob GF, Moal ML (1997) Drug abuse: hedonic homeostatic dysregulation. Science 278:52-58.

Koob GF, Moal ML (2006) Neurobiology of addiction. Academic Press, San Diego:69-120.

Kosten TA, Gawin FH, Kosten TR, Rounsaville BJ (1993) Gender differences in cocaine use and treatment response. J Subst Abuse Treat 10:63-66.

Kosten TR, Kosten TA, McDougle CJ, Hameedi FA, McCance EF, Rosen MI, Oliveto AH, Price LH (1996) Gender differences in response to intranasal cocaine administration to humans. Biol Psychiatry 39:147-148.

53

Kovacs K, Kiss JZ, Makara GB (1986) Glucocorticoid implants around the hypothalamic paraventricular nucleus prevent the increase of corticotropin-releasing factor and arginine vasopressin immunostaining induced by adrenalectomy. Neuroendocrinology 44:229-234.

Kreek M, Koob G (1998) Drug dependence: stress and dysregulation of brain reward pathways. Drug Alcohol Depend 51:23-47.

Krozowski ZS, Funder JW (1983) Renal mineralocorticoid receptors and hippocampal corticosterone-binding species have identical intrinsic steroid specificity. Proc Natl Acad Sci U S A 80:6056-6060.

Kudielka BM, Kirschbaum C (2005) Sex differences in HPA axis responses to stress: a review. Biol Psychol 69:113-132.

Kuhar MJ, Pilotte NS (1996) Neurochemical changes in cocaine withdrawal. Trends Pharmacol Sci 17:260-264.

Kuhn C, Francis R (1997) Gender difference in cocaine-induced HPA axis activation. Neuropsychopharmacology 16:399-407.

La Greca AM, Prinstein MJ, Fetter MD (2001) Adolescent peer crowd affiliation: linkages with health-risk behaviors and close friendships. J Pediatr Psychol 26:131-143.

Lamon BC, Alonzo A (1997) Stress among males recovering from substance abuse. Addict Behav 22:195-205.

Laviola G, Adriani W, Terranova ML, Gerra G (1999) Psychobiological risk factors for vulnerability to psychostimulants in human adolescents and animal models. Neuroscience and Biobehavioral Reviews 23:993-1010.

Laviola G, Wood RD, Kuhn C, Francis R, Spear LP (1995) Cocaine sensitization in periadolescent and adult rats. J Pharmacol Exp Ther 275:345-357.

Le Moal M, Simon H (1991) Mesocorticolimbic dopaminergic network: functional and regulatory roles. Physiol Rev 71:155-234.

Lepsch LB, Gonzalo LA, Magro FJ, Delucia R, Scavone C, Planeta CS (2005) Exposure to chronic stress increases the locomotor response to cocaine and the basal levels of corticosterone in adolescent rats. Addict Biol 10:251-256.

Lesniewska B, Nowak M, Malendowicz LK (1990a) Sex differences in adrenocortical structure and function. XXVIII. ACTH and corticosterone in intact, gonadectomised and gonadal hormone replaced rats. Horm Metab Res 22:378-381.

Lesniewska B, Miskowiak B, Nowak M, Malendowicz LK (1990b) Sex differences in adrenocortical structure and function. XXVII. The effect of ether stress on ACTH and corticosterone in intact, gonadectomized, and testosterone- or estradiol-replaced rats. Res Exp Med (Berl) 190:95-103.

Levy AD, Baumann MH, Van de Kar LD (1994a) Monoaminergic regulation of neuroendocrine function and its modification by cocaine. Front Neuroendocrinol 15:85-156.

Levy AD, Li Q, Alvarez Sanz MC, Rittenhouse PA, Kerr JE, Van de Kar LD (1992) Neuroendocrine responses to cocaine do not exhibit sensitization following repeated cocaine exposure. Life Sci 51:887-897.

Levy AD, Rittenhouse PA, Li Q, Yracheta J, Kunimoto K, Vandekar LD (1994b) Influence of repeated cocaine exposure on the endocrine and behavioral-responses to stress in rats. Psychopharmacology 113:547-554.

Levy AD, Li QA, Kerr JE, Rittenhouse PA, Milonas G, Cabrera TM, Battaglia G, Alvarez Sanz MC, Van de Kar LD (1991) Cocaine-induced elevation of plasma adrenocorticotropin hormone and corticosterone is mediated by serotonergic neurons. J Pharmacol Exp Ther 259:495-500.

Loh EA, Roberts DC (1990) Break-points on a progressive ratio schedule reinforced by intravenous cocaine increase following depletion of forebrain serotonin. Psychopharmacology (Berl) 101:262-266.

54

Lubman DI, Yucel M (2008) Adolescent substance use and the developing brain. Dev Med Child Neurol 50:76-77.

Lubman DI, Yucel M, Hall WD (2007) Substance use and the adolescent brain: a toxic combination? J Psychopharmacol 21:792-794.

Lucki I (1998) The spectrum of behaviors influenced by serotonin. Biol Psychiatry 44:151-162.

Luine VN (1993) Serotonin, catecholamines and metabolites in discrete brain areas in relation to lordotic responding on proestrus. Neuroendocrinology 57:946-954.

Lynch W, Carroll M (1999) Sex differences in the acquisition of intravenously self-administered cocaine and heroin in rats. Psychopharmacology (Berl) 144:77-82.

Lynch W, Roth M, Carroll M (2002) Biological basis of sex differences in drug abuse: preclinical and clinical studies. Psychopharmacology (Berl) 164:121-137.

Lynch W, Roth M, Mickelberg J, Carroll M (2001) Role of estrogen in the acquisition of intravenously self-administered cocaine in female rats. Pharmacol Biochem Behav 68:641-646.

Lynch WJ (2008) Acquisition and maintenance of cocaine self-administration in adolescent rats: effects of sex and gonadal hormones. Psychopharmacology (Berl) 197:237-246.

Lynch WJ, Carroll ME (2000) Reinstatement of cocaine self-administration in rats: sex differences. Psychopharmacology (Berl) 148:196-200.

Maeda K, Cagampang FR, Coen CW, Tsukamura H (1994) Involvement of the catecholaminergic input to the paraventricular nucleus and of corticotropin-releasing hormone in the fasting-induced suppression of luteinizing hormone release in female rats. Endocrinology 134:1718-1722.

Maggos CE, Spangler R, Zhou Y, Schlussman SD, Ho A, Kreek MJ (1997) Quantitation of dopamine transporter mRNA in the rat brain: mapping, effects of "binge" cocaine administration and withdrawal. Synapse 26:55-61.

Maggos CE, Tsukada H, Kakiuchi T, Nishiyama S, Myers JE, Kreuter J, Schlussman SD, Unterwald EM, Ho A, Kreek MJ (1998) Sustained withdrawal allows normalization of in vivo [11C]N-methylspiperone dopamine D2 receptor binding after chronic binge cocaine: a positron emission tomography study in rats. Neuropsychopharmacology 19:146-153.

Maisonneuve IM, Kreek MJ (1994) Acute tolerance to the dopamine response induced by a binge pattern of cocaine administration in male rats: an in vivo microdialysis study. J Pharmacol Exp Ther 268:916-921.

Maisonneuve IM, Ho A, Kreek MJ (1995) Chronic administration of a cocaine "binge" alters basal extracellular levels in male rats: an in vivo microdialysis study. J Pharmacol Exp Ther 272:652-657.

Maldonado AM, Kirstein CL (2005) Handling alters cocaine-induced activity in adolescent but not adult male rats. Physiol Behav 84:321-326.

Maldonado R, Robledo P, Chover AJ, Caine SB, Koob GF (1993) D1 dopamine receptors in the nucleus accumbens modulate cocaine self-administration in the rat. Pharmacol Biochem Behav 45:239-242.

Malendowicz LK, Mlynarczyk W (1982) Sex differences in adrenocortical structure and function. X. Lipid and corticosterone in the rat adrenal as affected by gonadectomy and testosterone or estradiol replacement. Endokrinologie 79:292-300.

Mantsch J, Goeders N (1999) Ketoconazole blocks the stress-induced reinstatement of cocaine-seeking behavior in rats: relationship to the discriminative stimulus effects of cocaine. Psychopharmacology (Berl) 142:399-407.

Mantsch JR, Saphier D, Goeders NE (1998) Corticosterone facilitates the acquisition of cocaine self-administration in rats: Opposite effects of the type II glucocorticoid receptor agonist dexamethasone. J Pharmacol Exp Ther 287:72-80.

55

Mantsch JR, Schlussman SD, Ho A, Kreek MJ (2000) Effects of cocaine self-administration on plasma corticosterone and prolactin in rats. J Pharmacol Exp Ther 294:239-247.

Mantsch JR, Yuferov V, Mathieu-Kia AM, Ho A, Kreek MJ (2003) Neuroendocrine alterations in a high-dose, extended-access rat self-administration model of escalating cocaine use. Psychoneuroendocrinology 28:836-862.

Marin MT, Cruz FC, Planeta CS (2008) Cocaine-induced behavioral sensitization in adolescent rats endures until adulthood: Lack of association with GluR1 and NR1 glutamate receptor subunits and tyrosine hydroxylase. Pharmacol Biochem Behav 91:109-114.

Marinelli M, Piazza P (2002) Interaction between glucocorticoid hormones, stress and psychostimulant drugs. Eur J Neurosci 16:387-394.

Martin-Iverson MT, Szostak C, Fibiger HC (1986) 6-Hydroxydopamine lesions of the medial prefrontal cortex fail to influence intravenous self-administration of cocaine. Psychopharmacology (Berl) 88:310-314.

Martin CA, Kelly TH, Rayens MK, Brogli BR, Brenzel A, Smith WJ, Omar HA (2002) Sensation seeking, puberty, and nicotine, alcohol, and marijuana use in adolescence. J Am Acad Child Adolesc Psychiatry 41:1495-1502.

Mash D, Staley J, Izenwasser S, Basile M, Ruttenber A (2000) Serotonin transporters upregulate with chronic cocaine use. J Chem Neuroanat 20:271-280.

Matell MS, King GR (1997) 5-HT3 receptor mediated dopamine release in the nucleus accumbens during withdrawal from continuous cocaine. Psychopharmacology (Berl) 130:242-248.

McCance-Katz EF, Carroll KM, Rounsaville BJ (1999) Gender differences in treatment-seeking cocaine abusers--implications for treatment and prognosis. Am J Addict 8:300-311.

McCance-Katz EF, Hart CL, Boyarsky B, Kosten T, Jatlow P (2005) Gender effects following repeated administration of cocaine and alcohol in humans. Subst Use Misuse 40:511-528.

McCormick CM, Mathews IZ (2007) HPA function in adolescence: role of sex hormones in its regulation and the enduring consequences of exposure to stressors. Pharmacol Biochem Behav 86:220-233.

McCormick CM, Smith C, Mathews IZ (2008) Effects of chronic social stress in adolescence on anxiety and neuroendocrine response to mild stress in male and female rats. Behav Brain Res 187:228-238.

McCormick CM, Kehoe P, Mallinson K, Cecchi L, Frye CA (2002) Neonatal isolation alters stress hormone and mesolimbic dopamine release in juvenile rats. Pharmacol Biochem Behav 73:77-85.

McEwen BS (1980) Steroid hormones and the brain: cellular mechanisms underlying neural and behavioral plasticity. Psychoneuroendocrinology 5:1-11.

McEwen BS (2000) The neurobiology of stress: from serendipity to clinical relevance. Brain Res 886:172-189.

McQueen JK, Wilson H, Sumner BE, Fink G (1999) Serotonin transporter (SERT) mRNA and binding site densities in male rat brain affected by sex steroids. Brain Res Mol Brain Res 63:241-247.

Meil WM, See RE (1996) Conditioned cued recovery of responding following prolonged withdrawal from self-administered cocaine in rats: an animal model of relapse. Behav Pharmacol 7:754-763.

Mello N, Mendelson J (1997) Cocaine's effects on neuroendocrine systems: clinical and preclinical studies. Pharmacol Biochem Behav 57:571-599.

Mello NK, Mendelson JH (2002) Cocaine, hormones, and behavior: Clinical and preclinical studies. In: Hormones, Brain and Behavior (Pfaff DW, Arnold AP, Etgen AM, Fahrbach SE, Rubin RT, eds), pp 665-686. San Diego, California: Academic Press.

56

Mello NK, Mendelson JH, Drieze J, Kelly M (1990) Acute effects of cocaine on prolactin and gonadotropins in female rhesus monkey during the follicular phase of the menstrual cycle. J Pharmacol Exp Ther 254:815-823.

Mello NK, Mendelson JH, Kelly M, Bowen CA (2000) The effects of cocaine on basal and human chorionic gonadotropin-stimulated ovarian steroid hormones in female rhesus monkeys. J Pharmacol Exp Ther 294:1137-1145.

Mello NK, Sarnyai Z, Mendelson JH, Drieze JM, Kelly M (1993) Acute effects of cocaine on anterior pituitary hormones in male and female rhesus monkeys. J Pharmacol Exp Ther 266:804-811.

Mello NK, Mendelson JH, Kelly M, Diaz-Migoyo N, Sholar JW (1997) The Effects of Chronic Cocaine Self-Administration on the Menstrual Cycle in Rhesus Monkeys. J Pharmacol Exp Ther 281:70-83.

Mendelson J, Mello N, Sholar M, Siegel A, Mutschler N, Halpern J (2002) Temporal concordance of cocaine effects on mood states and neuroendocrine hormones. Psychoneuroendocrinology 27:71-82.

Mendelson J, Mello N, Sholar M, Siegel A, Kaufman M, Levin J, Renshaw P, Cohen B (1999a) Cocaine pharmacokinetics in men and in women during the follicular and luteal phases of the menstrual cycle. Neuropsychopharmacology 21:294-303.

Mendelson JH, Mello NK, Negus SS (1999b) Effects of luteinizing hormone-releasing hormone on plasma cocaine levels in rhesus monkeys. J Pharmacol Exp Ther 289:791-799.

Mendelson JH, Sholar MB, Siegel AJ, Mello NK (2001) Effects of cocaine on luteinizing hormone in women during the follicular and luteal phases of the menstrual cycle and in men. J Pharmacol Exp Ther 296:972-979.

Mendelson JH, Mello NK, Teoh SK, Ellingboe J, Cochin J (1989) Cocaine effects on pulsatile secretion of anterior pituitary, gonadal, and adrenal hormones. J Clin Endocrinol Metab 69:1256-1260.

Mendelson JH, Sholar M, Mello NK, Teoh SK, Sholar JW (1998) Cocaine tolerance: behavioral, cardiovascular, and neuroendocrine function in men. Neuropsychopharmacology 18:263-271.

Mendelson JH, Sholar MB, Mutschler NH, Jaszyna-Gasior M, Goletiani NV, Siegel AJ, Mello NK (2003) Effects of intravenous cocaine and cigarette smoking on luteinizing hormone, testosterone, and prolactin in men. J Pharmacol Exp Ther 307:339-348.

Merens W, Willem Van der Does AJ, Spinhoven P (2007) The effects of serotonin manipulations on emotional information processing and mood. J Affect Disord 103:43-62.

Miczek KA, Mutschler NH (1996) Activational effects of social stress on IV cocaine self-administration in rats. Psychopharmacology (Berl) 128:256-264.

Miller NS, Summers GL, Gold MS (1993) Cocaine dependence: alcohol and other drug dependence and withdrawal characteristics. J Addict Dis 12:25-35.

Mogenson GJ, Jones DL, Yim CY (1980) From motivation to action: functional interface between the limbic system and the motor system. Prog Neurobiol 14:69-97.

Moore RJ, Vinsant SL, Nader MA, Porrino LJ, Friedman DP (1998a) Effect of cocaine self-administration on dopamine D2 receptors in rhesus monkeys. Synapse 30:88-96.

Moore RJ, Vinsant SL, Nader MA, Porrino LJ, Friedman DP (1998b) Effect of cocaine self-administration on striatal dopamine D1 receptors in rhesus monkeys. Synapse 28:1-9.

Mountjoy KG, Robbins LS, Mortrud MT, Cone RD (1992) The cloning of a family of genes that encode the melanocortin receptors. Science 257:1248-1251.

Muller CP, Huston JP (2006) Determining the region-specific contributions of 5-HT receptors to the psychostimulant effects of cocaine. Trends Pharmacol Sci 27:105-112.

Muller CP, Carey RJ, Huston JP (2003) Serotonin as an important mediator of cocaine's behavioral effects. Drugs Today (Barc) 39:497-511.

57

Muller CP, Carey RJ, De Souza Silva MA, Jocham G, Huston JP (2002a) Cocaine increases serotonergic activity in the hippocampus and nucleus accumbens in vivo: 5-HT1a-receptor antagonism blocks behavioral but potentiates serotonergic activation. Synapse 45:67-77.

Muller CP, De Souza Silva MA, DePalma G, Tomaz C, Carey RJ, Huston JP (2002b) The selective serotonin(1A)-receptor antagonist WAY 100635 blocks behavioral stimulating effects of cocaine but not ventral striatal dopamine increase. Behav Brain Res 134:337-346.

Musselman DL, Nemeroff CB (1996) Depression and endocrine disorders: focus on the thyroid and adrenal system. Br J Psychiatry Suppl:123-128.

Mutiara S, Kanasaki H, Harada T, Miyazaki K (2006) Dopamine D(2) receptor expression and regulation of gonadotropin alpha-subunit gene in clonal gonadotroph LbetaT2 cells. Mol Cell Endocrinol 259:22-29.

Nemeroff CB (1998) Psychopharmacology of affective disorders in the 21st century. Biol Psychiatry 44:517-525.

Nestler E (2001) Molecular basis of long-term plasticity underlying addiction. Nat Rev Neurosci 2:119-128.

Neumaier JF, Vincow ES, Arvanitogiannis A, Wise RA, Carlezon WA, Jr. (2002) Elevated expression of 5-HT1B receptors in nucleus accumbens efferents sensitizes animals to cocaine. J Neurosci 22:10856-10863.

Nippoldt TB, Reame NE, Kelch RP, Marshall JC (1989) The roles of estradiol and progesterone in decreasing luteinizing hormone pulse frequency in the luteal phase of the menstrual cycle. J Clin Endocrinol Metab 69:67-76.

Norman AW, Litwack G (1998) Hormones, Second Edition. San Diego, California: Academic press.

Norman RL, Smith CJ, Pappas JD, Hall J (1992) Exposure to ovarian steroids elicits a female pattern of plasma cortisol levels in castrated male macaques. Steroids 57:37-43.

Nowak KW, Neri G, Nussdorfer GG, Malendowicz LK (1995) Effects of sex hormones on the steroidogenic activity of dispersed adrenocortical cells of the rat adrenal cortex. Life Sci 57:833-837.

O'Neill MF, Heron-Maxwell CL, Shaw G (1999) 5-HT2 receptor antagonism reduces hyperactivity induced by amphetamine, cocaine, and MK-801 but not D1 agonist C-APB. Pharmacol Biochem Behav 63:237-243.

Oades RD, Halliday GM (1987) Ventral tegmental (A10) system: neurobiology. 1. Anatomy and connectivity. Brain Res 434:117-165.

Olster DH, Ferin M (1987) Corticotropin-releasing hormone inhibits gonadotropin secretion in the ovariectomized rhesus monkey. J Clin Endocrinol Metab 65:262-267.

Papasava M, Singer G (1985) Self-administration of low-dose cocaine by rats at reduced and recovered body weight. Psychopharmacology (Berl) 85:419-425.

Parsons LH, Koob GF, Weiss F (1995) Serotonin dysfunction in the nucleus accumbens of rats during withdrawal after unlimited access to intravenous cocaine. J Pharmacol Exp Ther 274:1182-1191.

Parsons LH, Koob GF, Weiss F (1996a) Extracellular serotonin is decreased in the nucleus accumbens during withdrawal from cocaine self-administration. Behav Brain Res 73:225-228.

Parsons LH, Weiss F, Koob GF (1996b) Serotonin1b receptor stimulation enhances dopamine-mediated reinforcement. Psychopharmacology (Berl) 128:150-160.

Payne AH, Kelch RP, Murono EP, Kerlan JT (1977) Hypothalamic, pituitary and testicular function during sexual maturation of the male rat. J Endocrinol 72:17-26.

Peltier R, Schenk S (1993) Effects of serotonergic manipulations on cocaine self-administration in rats. Psychopharmacology (Berl) 110:390-394.

58

Perry JL, Anderson MM, Nelson SE, Carroll ME (2007) Acquisition of i.v. cocaine self-administration in adolescent and adult male rats selectively bred for high and low saccharin intake. Physiol Behav 91:126-133.

Petraglia F, Sutton S, Vale W, Plotsky P (1987) Corticotropin-releasing factor decreases plasma luteinizing hormone levels in female rats by inhibiting gonadotropin-releasing hormone release into hypophysial-portal circulation. Endocrinology 120:1083-1088.

Pettit HO, Justice JB, Jr. (1989) Dopamine in the nucleus accumbens during cocaine self-administration as studied by in vivo microdialysis. Pharmacol Biochem Behav 34:899-904.

Pettit HO, Ettenberg A, Bloom FE, Koob GF (1984) Destruction of dopamine in the nucleus accumbens selectively attenuates cocaine but not heroin self-administration in rats. Psychopharmacology (Berl) 84:167-173.

Philpot RM, Kirstein CL (1999) Repeated cocaine exposure: effects on catecholamines in the nucleus accumbens septi of periadolescent animals. Pharmacol Biochem Behav 62:465-472.

Piazza P, Le Moal M (1996) Pathophysiological basis of vulnerability to drug abuse: role of an interaction between stress, glucocorticoids, and dopaminergic neurons. Annu Rev Pharmacol Toxicol 36:359-378.

Piazza P, Le Moal M (1998) The role of stress in drug self-administration. Trends Pharmacol Sci 19:67-74.

Piazza PV, Le Moal M (1997) Glucocorticoids as a biological substrate of reward: physiological and pathophysiological implications. Brain Res Brain Res Rev 25:359-372.

Piazza PV, Deminiere JM, le Moal M, Simon H (1990) Stress- and pharmacologically-induced behavioral sensitization increases vulnerability to acquisition of amphetamine self-administration. Brain Res 514:22-26.

Piazza PV, Rouge-Pont F, Deroche V, Maccari S, Simon H, Le Moal M (1996) Glucocorticoids have state-dependent stimulant effects on the mesencephalic dopaminergic transmission. PNAS 93:8716-8720.

Piazza PV, Marinelli M, Jodogne C, Deroche V, Rouge-Pont F, Maccari S, Le Moal M, Simon H (1994) Inhibition of corticosterone synthesis by Metyrapone decreases cocaine-induced locomotion and relapse of cocaine self-administration. Brain Res 658:259-264.

Pierce R, Kalivas P (1997a) Repeated cocaine modifies the mechanism by which amphetamine releases dopamine. Journal of Neuroscience 17:3254-3261.

Pierce RC, Kalivas PW (1997b) A circuitry model of the expression of behavioral sensitization to amphetamine-like psychostimulants. Brain Res Brain Res Rev 25:192-216.

Pollack MH, Rosenbaum JF (1991) Fluoxetine treatment of cocaine abuse in heroin addicts. J Clin Psychiatry 52:31-33.

Porrino LJ, Ritz MC, Goodman NL, Sharpe LG, Kuhar MJ, Goldberg SR (1989) Differential effects of the pharmacological manipulation of serotonin systems on cocaine and amphetamine self-administration in rats. Life Sci 45:1529-1535.

Post RM, Weiss SR, Fontana D, Pert A (1992) Conditioned sensitization to the psychomotor stimulant cocaine. Ann N Y Acad Sci 654:386-399.

Prasad B, Ulibarri C, Sorg B (1998) Stress-induced cross-sensitization to cocaine: effect of adrenalectomy and corticosterone after short- and long-term withdrawal. Psychopharmacology (Berl) 136:24-33.

Przegalinski E, Filip M, Siwanowicz J, Nowak E (2000) Effect of adrenalectomy and corticosterone on cocaine-induced sensitization in rats. J Physiol Pharmacol 51:193-204.

Quinones-Jenab V, Ho A, Schlussman SD, Franck J, Kreek MJ (1999) Estrous cycle differences in cocaine-induced stereotypic and locomotor behaviors in Fischer rats. Behav Brain Res 101:15-20.

59

Quinones-Jenab V, Krey LC, Schlussman SD, Ho A, Kreek MJ (2000a) Chronic 'binge' pattern cocaine alters the neuroendocrine profile of pregnant rats. Neurosci Lett 282:120-122.

Quinones-Jenab V, Perrotti LI, Fabian SJ, Chin J, Russo SJ, Jenab S (2001) Endocrinological basis of sex differences in cocaine-induced behavioral responses. Ann NY Acad Sci 937:140-171.

Quinones-Jenab V, Perrotti LI, Ho A, Jenab S, Schlussman SD, Franck J, Kreek MJ (2000b) Cocaine affects progesterone plasma levels in female rats. Pharmacol Biochem Behav 66:449-453.

Raap D, Morin B, Medici C, Smith R (2000) Adolescent cocaine and injection stress effects on the estrous cycle. Physiol Behav 70:417-424.

Rapoport JL, Giedd JN, Blumenthal J, Hamburger S, Jeffries N, Fernandez T, Nicolson R, Bedwell J, Lenane M, Zijdenbos A, Paus T, Evans A (1999) Progressive cortical change during adolescence in childhood-onset schizophrenia. A longitudinal magnetic resonance imaging study. Arch Gen Psychiatry 56:649-654.

Reilly JG, McTavish SF, Young AH (1997) Rapid depletion of plasma tryptophan: a review of studies and experimental methodology. J Psychopharmacol 11:381-392.

Reith ME (1990) 5-HT3 receptor antagonists attenuate cocaine-induced locomotion in mice. Eur J Pharmacol 186:327-330.

Ritz MC, Cone EJ, Kuhar MJ (1990) Cocaine inhibition of ligand binding at dopamine, norepinephrine and serotonin transporters: a structure-activity study. Life Sci 46:635-645.

Ritz MC, Lamb RJ, Goldberg SR, Kuhar MJ (1987) Cocaine receptors on dopamine transporters are related to self-administration of cocaine. Science 237:1219-1223.

Rivier C, Vale W (1987) Cocaine stimulates adrenocorticotropin (ACTH) secretion through a corticotropin-releasing factor (CRF)-mediated mechanism. Brain Res 422:403-406.

Rivier C, Rivier J, Vale W (1986) Stress-induced inhibition of reproductive functions: role of endogenous corticotropin-releasing factor. Science 231:607-609.

Robbins SJ, Ehrman RN, Childress AR, O'Brien CP (1999) Comparing levels of cocaine cue reactivity in male and female outpatients. Drug Alcohol Depend 53:223-230.

Robbins TW, Everitt BJ (1996) Neurobehavioural mechanisms of reward and motivation. Curr Opin Neurobiol 6:228-236.

Roberts DC, Koob GF (1982) Disruption of cocaine self-administration following 6-hydroxydopamine lesions of the ventral tegmental area in rats. Pharmacol Biochem Behav 17:901-904.

Roberts DC, Koob GF, Klonoff P, Fibiger HC (1980) Extinction and recovery of cocaine self-administration following 6-hydroxydopamine lesions of the nucleus accumbens. Pharmacol Biochem Behav 12:781-787.

Robinson TE, Berridge KC (2003) Addiction. Annu Rev Psychol 54:25-53.

Rocha BA, Fumagalli F, Gainetdinov RR, Jones SR, Ator R, Giros B, Miller GW, Caron MG (1998) Cocaine self-administration in dopamine-transporter knockout mice. Nat Neurosci 1:132-137.

Rogerio, Takahashi RN (1992) Anxiogenic properties of cocaine in the rat evaluated with the elevated plus-maze. Pharmacol Biochem Behav 43:631-633.

Romach MK, Glue P, Kampman K, Kaplan HL, Somer GR, Poole S, Clarke L, Coffin V, Cornish J, O'Brien CP, Sellers EM (1999) Attenuation of the euphoric effects of cocaine by the dopamine D1/D5 antagonist ecopipam (SCH 39166). Arch Gen Psychiatry 56:1101-1106.

Rosenberg DR, Lewis DA (1994) Changes in the dopaminergic innervation of monkey prefrontal cortex during late postnatal development: a tyrosine hydroxylase immunohistochemical study. Biol Psychiatry 36:272-277.

Rosner W (1990) The functions of corticosteroid-binding globulin and sex hormone-binding globulin: recent advances. Endocr Rev 11:80-91.

60

Rothman RB, Baumann MH, Dersch CM, Romero DV, Rice KC, Carroll FI, Partilla JS (2001) Amphetamine-type central nervous system stimulants release norepinephrine more potently than they release dopamine and serotonin. Synapse 39:32-41.

Rothman RB, Blough BE, Woolverton WL, Anderson KG, Negus SS, Mello NK, Roth BL, Baumann MH (2005) Development of a rationally designed, low abuse potential, biogenic amine releaser that suppresses cocaine self-administration. J Pharmacol Exp Ther 313:1361-1369.

Rouge-Pont F, Marinelli M, Le Moal M, Simon H, Piazza PV (1995) Stress-induced sensitization and glucocorticoids. II. Sensitization of the increase in extracellular dopamine induced by cocaine depends on stress-induced corticosterone secretion. J Neurosci 15:7189-7195.

Rupprecht R, Reul JM, van Steensel B, Spengler D, Soder M, Berning B, Holsboer F, Damm K (1993) Pharmacological and functional characterization of human mineralocorticoid and glucocorticoid receptor ligands. Eur J Pharmacol 247:145-154.

Russo S, Jenab S, Fabian S, Festa E, Kemen L, Quinones-Jenab V (2003a) Sex differences in the conditioned rewarding effects of cocaine. Brain Research 970:214-220.

Russo S, Festa E, Fabian S, Gazi F, Kraish M, Jenab S, Quinones-Jenab V (2003b) Gonadal hormones differentially modulate cocaine-induced conditioned place preference in male and female rats. Neuroscience 120:523-533.

Sanchez MM, Young LJ, Plotsky PM, Insel TR (2000) Distribution of corticosteroid receptors in the rhesus brain: relative absence of glucocorticoid receptors in the hippocampal formation. J Neurosci 20:4657-4668.

Santucci AC (2008) Adolescent cocaine residually impairs working memory and enhances fear memory in rats. Exp Clin Psychopharmacol 16:77-85.

Santucci AC, Madeira E (2008) Anxiogenesis in adult rats treated chronically with cocaine during adolescence: effects of extended abstinence and 8-OH-DPAT treatment. Brain Res Bull 76:402-411.

Santucci AC, Capodilupo S, Bernstein J, Gomez-Ramirez M, Milefsky R, Mitchell HU- (2004) Cocaine in adolescent rats produces residual memory impairments that are reversible with time. Neurotoxicology and Teratology 26:651-661.

Saphier D, Goeders NE, Welch JE, Farrar GE (1993) Effects of intracerebroventricular and intrahypothalamic cocaine administration on adrenocortical secretion. Neuroendocrinology 57:54-62.

Sarnyai Z, Shaham Y, Heinrichs SC (2001) The role of corticotropin-releasing factor in drug addiction. Pharmacol Rev 53:209-244.

Sarnyai Z, Biro E, Penke B, Telegdy G (1992) The cocaine-induced elevation of plasma corticosterone is mediated by endogenous corticotropin-releasing factor (CRF) in rats. Brain Res 589:154-156.

Sarnyai Z, Mello NK, Mendelson JH, Eros-Sarnyai M, Mercer G (1996) Effects of cocaine on pulsatile activity of hypothalamic-pituitary-adrenal axis in male rhesus monkeys: neuroendocrine and behavioral correlates. J Pharmacol Exp Ther 277:225-234.

Sarnyai Z, Biro E, Gardi J, Vecsernyes M, Julesz J, Telegdy G (1993) Alterations of corticotropin-releasing factor-like immunoreactivity in different brain regions after acute cocaine administration in rats. Brain Res 616:315-319.

Sarnyai Z, Vecsernyés M, Julesz J, Bíró E, Gardi J, Telegdy G (1995) Brain corticotropin-releasing factor mediates 'anxiety-like' behavior induced by cocaine withdrawal in rats. Brain Res 675:89-97.

Satel SL, Price LH, Palumbo JM, McDougle CJ, Krystal JH, Gawin F, Charney DS, Heninger GR, Kleber HD (1991) Clinical phenomenology and neurobiology of cocaine abstinence: a prospective inpatient study. Am J Psychiatry 148:1712-1716.

Schmidt ED, Tilders FJ, Janszen AW, Binnekade R, De Vries TJ, Schoffelmeer AN (1995) Intermittent cocaine exposure causes delayed and long-lasting sensitization of cocaine-induced ACTH secretion in rats. Eur J Pharmacol 285:317-321.

61

Schramm-Sapyta NL, Morris RW, Kuhn CM (2006) Adolescent rats are protected from the conditioned aversive properties of cocaine and lithium chloride. Pharmacology Biochemistry and Behavior 84:344-352.

Seale JV, Wood SA, Atkinson HC, Harbuz MS, Lightman SL (2004a) Gonadal steroid replacement reverses gonadectomy-induced changes in the corticosterone pulse profile and stress-induced hypothalamic-pituitary-adrenal axis activity of male and female rats. Journal of Neuroendocrinology 16:989-998.

Seale JV, Wood SA, Atkinson HC, Bate E, Lightman SL, Ingram CD, Jessop DS, Harbuz MS (2004b) Gonadectomy reverses the sexually diergic patterns of circadian and stress-induced hypothalamic-pituitary-adrenal axis activity in male and female rats. J Neuroendocrinol 16:516-524.

See RE (2002) Neural substrates of conditioned-cued relapse to drug-seeking behavior. Pharmacol Biochem Behav 71:517-529.

Sell SL, Scalzitti JM, Thomas ML, Cunningham KA (2000) Influence of ovarian hormones and estrous cycle on the behavioral response to cocaine in female rats. J Pharmacol Exp Ther 293:879-886.

Sellings LH, McQuade LE, Clarke PB (2006) Evidence for multiple sites within rat ventral striatum mediating cocaine-conditioned place preference and locomotor activation. J Pharmacol Exp Ther 317:1178-1187.

Shaham Y, Erb S, Stewart J (2000) Stress-induced relapse to heroin and cocaine seeking in rats: a review. Brain Research Reviews 33:13-33.

Shaham Y, Erb S, Leung S, Buczek Y, Stewart J (1998) CP-154,526, a selective, non-peptide antagonist of the corticotropin-releasing factor1 receptor attenuates stress-induced relapse to drug seeking in cocaine- and heroin-trained rats. Psychopharmacology (Berl) 137:184-190.

Shalev U, Marinelli M, Baumann MH, Piazza PV, Shaham Y (2003) The role of corticosterone in food deprivation-induced reinstatement of cocaine seeking in the rat. Psychopharmacology (Berl) 168:170-176.

Sholar MB, Mendelson JH, Mello NK, Siegel AJ, Kaufman MJ, Levin JM, Renshaw PF, Cohen BM (1998) Concurrent pharmacokinetic analysis of plasma cocaine and adrenocorticotropic hormone in men. J Clin Endocrinol Metab 83:966-968.

Sinha R (2001) How does stress increase risk of drug abuse and relapse? Psychopharmacology (Berl) 158:343-359.

Sinha R, Fuse T, Aubin L, O'Malley S (2000) Psychological stress, drug-related cues and cocaine craving. Psychopharmacology (Berl) 152:140-148.

Sisk CL, Foster DL (2004) The neural basis of puberty and adolescence. Nat Neurosci 7:1040-1047.

Sisk CL, Zehr JL (2005) Pubertal hormones organize the adolescent brain and behavior. Front Neuroendocrinol 26:163-174.

Smith DE, Wesson DR, Apter-Marsh M (1984) Cocaine- and alcohol-induced sexual dysfunction in patients with addictive disease. J Psychoactive Drugs 16:359-361.

Sofuoglu M, Dudish-Poulsen S, Nelson D, Pentel PR, Hatsukami DK (1999) Sex and menstrual cycle differences in the subjective effects from smoked cocaine in humans. Exp Clin Psychopharmacol 7:274-283.

Sora I, Hall FS, Andrews AM, Itokawa M, Li XF, Wei HB, Wichems C, Lesch KP, Murphy DL, Uhl GR (2001) Molecular mechanisms of cocaine reward: combined dopamine and serotonin transporter knockouts eliminate cocaine place preference. Proc Natl Acad Sci U S A 98:5300-5305.

Sowell ER, Peterson BS, Thompson PM, Welcome SE, Henkenius AL, Toga AW (2003) Mapping cortical change across the human life span. Nat Neurosci 6:309-315.

62

Spear L (2000) The adolescent brain and age-related behavioral manifestations. Neurosci Biobehav Rev 24:417-463.

Spear LP (2007) Assessment of adolescent neurotoxicity: rationale and methodological considerations. Neurotoxicol Teratol 29:1-9.

Spear LP, Silveri MM, Casale M, Katovic NM, Campbell JO, Douglas LA (2002) Cocaine and development: A retrospective perspective. Neurotoxicology and Teratology 24:321-327.

Spyraki C, Fibiger HC, Phillips AG (1982) Cocaine-induced place preference conditioning: lack of effects of neuroleptics and 6-hydroxydopamine lesions. Brain Res 253:195-203.

Stansfield KH, Kirstein CL (2005) Neurochemical effects of cocaine in adolescence compared to adulthood. Brain Res Dev Brain Res 159:119-125.

Stansfield KH, Kirstein CL (2006) Effects of novelty on behavior in the adolescent and adult rat. Dev Psychobiol 48:10-15.

Stansfield KH, Kirstein CL (2007) Chronic cocaine or ethanol exposure during adolescence alters novelty-related behaviors in adulthood. Pharmacol Biochem Behav 86:637-642.

Swerdloff RS, Wang C, Hikim APS (2002) Hypothalamic-pituitary-gonadal axis in men. In: Hormones, Brain and Behavior (Pfaff DW, Arnold AP, Etgen AM, Fahrbach SE, Rubin RT, eds), pp 1-16. San Diego, California: Academic Press.

Szumlinski KK, Frys KA, Kalivas PW (2004) Dissociable roles for the dorsal and median raphe in the facilitatory effect of 5-HT1A receptor stimulation upon cocaine-induced locomotion and sensitization. Neuropsychopharmacology 29:1675-1687.

Teicher MH, Barber NI, Gelbard HA, Gallitano AL, Campbell A, Marsh E, Baldessarini RJ (1993) Developmental differences in acute nigrostriatal and mesocorticolimbic system response to haloperidol. Neuropsychopharmacology 9:147-156.

Teitsma CA, Anglade I, Lethimonier C, Le Drean G, Saligaut D, Ducouret B, Kah O (1999) Glucocorticoid receptor immunoreactivity in neurons and pituitary cells implicated in reproductive functions in rainbow trout: a double immunohistochemical study. Biol Reprod 60:642-650.

Teoh SK, Lex BW, Mendelson JH, Mello NK, Cochin J (1992) Hyperprolactinemia and macrocytosis in women with alcohol and polysubstance dependence. J Stud Alcohol 53:176-182.

Teoh SK, Sarnyai Z, Mendelson JH, Mello NK, Springer SA, Sholar JW, Wapler M, Kuehnle JC, Gelles H (1994) Cocaine effects on pulsatile secretion of ACTH in men. J Pharmacol Exp Ther 270:1134-1138.

Thompson TL, Moss RL (1994) Estrogen regulation of dopamine release in the nucleus accumbens: genomic- and nongenomic-mediated effects. J Neurochem 62:1750-1756.

Thompson TL, Moore CC, Smith B (2000) Estrogen priming modulates autoreceptor-mediated potentiation of dopamine uptake. Eur J Pharmacol 401:357-363.

Tidey J, Miczek K (1997) Acquisition of cocaine self-administration after social stress: role of accumbens dopamine. Psychopharmacology (Berl) 130:203-212.

Tinnikov AA (1999) Responses of serum corticosterone and corticosteroid-binding globulin to acute and prolonged stress in the rat. Endocrine 11:145-150.

Torres G, Rivier C (1992a) Cocaine-induced ACTH secretion: dependence of plasma levels of the drug and mode of exposure. Brain Res Bull 29:51-56.

Torres G, Rivier C (1992b) Differential effects of intermittent or continuous exposure to cocaine on the hypothalamic-pituitary-adrenal axis and c-fos expression. Brain Res 571:204-211.

Tsukada H, Kreuter J, Maggos CE, Unterwald EM, Kakiuchi T, Nishiyama S, Futatsubashi M, Kreek MJ (1996) Effects of binge pattern cocaine administration on dopamine D1 and D2 receptors in the rat brain: An in vivo study using positron emission tomography. J Neurosci 16:7670-7677.

63

UNODC (2008) 2008 Word drug report. In, pp 1-96: Office of Drug and Crime of the United Nations.

Vamvakopoulos NC, Chrousos GP (1993) Evidence of direct estrogenic regulation of human corticotropin-releasing hormone gene expression. Potential implications for the sexual dimophism of the stress response and immune/inflammatory reaction. J Clin Invest 92:1896-1902.

Van Bockstaele EJ, Cestari DM, Pickel VM (1994) Synaptic structure and connectivity of serotonin terminals in the ventral tegmental area: potential sites for modulation of mesolimbic dopamine neurons. Brain Res 647:307-322.

van Luijtelaar E, Dirksen R, Vree T, van Haaren F (1996) Effects of acute and chronic cocaine administration on EEG and behaviour in intact and castrated male and intact and ovariectomized female rats. Brain Res Bull 40:43-50.

Vescovi PP, Coiro V, Volpi R, Passeri M (1992) Diurnal variations in plasma ACTH, cortisol and beta-endorphin levels in cocaine addicts. Horm Res 37:221-224.

Viau V, Meaney MJ (1991) Variations in the hypothalamic-pituitary-adrenal response to stress during the estrous cycle in the rat. Endocrinology 129:2503-2511.

Viau V, Meaney MJ (1996) The inhibitory effect of testosterone on hypothalamic-pituitary-adrenal responses to stress is mediated by the medial preoptic area. J Neurosci 16:1866-1876.

Volkow ND, Fowler JS, Wang GJ (1999) Imaging studies on the role of dopamine in cocaine reinforcement and addiction in humans. J Psychopharmacol 13:337-345.

Volkow ND, Fowler JS, Wang G-J (2003) The addicted human brain: insights from imaging studies. J Clin Invest 111:1444-1451.

Volkow ND, Fowler JS, Wang GJ, Hitzemann R, Logan J, Schlyer DJ, Dewey SL, Wolf AP (1993) Decreased dopamine D2 receptor availability is associated with reduced frontal metabolism in cocaine abusers. Synapse 14:169-177.

Walker QD, Kuhn CM (2008) Cocaine increases stimulated dopamine release more in periadolescent than adult rats. Neurotoxicol Teratol 30:412-418.

Walker QD, Rooney MB, Wightman RM, Kuhn CM (2000) Dopamine release and uptake are greater in female than male rat striatum as measured by fast cyclic voltammetry. Neuroscience 95:1061-1070.

Walker QD, Francis R, Cabassa J, Kuhn CM (2001a) Effect of ovarian hormones and estrous cycle on stimulation of the hypothalamo-pituitary-adrenal axis by cocaine. J Pharmacol Exp Ther 297:291-298.

Walker QD, Cabassa J, Kaplan KA, Li ST, Haroon J, Spohr HA, Kuhn CM (2001b) Sex differences in cocaine-stimulated motor behavior: disparate effects of gonadectomy. Neuropsychopharmacology 25:118-130.

Warner LA, Kessler RC, Hughes M, Anthony JC, Nelson CB (1995) Prevalence and correlates of drug use and dependence in the United States. Results from the National Comorbidity Survey. Arch Gen Psychiatry 52:219-229.

Watts AG, Swanson LW (1987) Efferent projections of the suprachiasmatic nucleus: II. Studies using retrograde transport of fluorescent dyes and simultaneous peptide immunohistochemistry in the rat. J Comp Neurol 258:230-252.

Watts AG, Swanson LW (1989) Diurnal variations in the content of preprocorticotropin-releasing hormone messenger ribonucleic acids in the hypothalamic paraventricular nucleus of rats of both sexes as measured by in situ hybridization. Endocrinology 125:1734-1738.

Weddington WW, Brown BS, Haertzen CA, Cone EJ, Dax EM, Herning RI, Michaelson BS (1990) Changes in mood, craving, and sleep during short-term abstinence reported by male cocaine addicts. A controlled, residential study. Arch Gen Psychiatry 47:861-868.

Wee S, Anderson KG, Baumann MH, Rothman RB, Blough BE, Woolverton WL (2005) Relationship between the serotonergic activity and reinforcing effects of a series of amphetamine analogs. J Pharmacol Exp Ther 313:848-854.

64

Weiss F, Markou A, Lorang MT, Koob GF (1992) Basal extracellular dopamine levels in the nucleus accumbens are decreased during cocaine withdrawal after unlimited-access self-administration. Brain Res 593:314-318.

Wiemann JN, Clifton DK, Steiner RA (1989) Pubertal changes in gonadotropin-releasing hormone and proopiomelanocortin gene expression in the brain of the male rat. Endocrinology 124:1760-1767.

Wills TA, Vaccaro D, McNamara G (1994) Novelty seeking, risk taking, and related constructs as predictors of adolescent substance use: an application of Cloninger's theory. J Subst Abuse 6:1-20.

Woo TU, Pucak ML, Kye CH, Matus CV, Lewis DA (1997) Peripubertal refinement of the intrinsic and associational circuitry in monkey prefrontal cortex. Neuroscience 80:1149-1158.

Wu JC, Bell K, Najafi A, Widmark C, Keator D, Tang C, Klein E, Bunney BG, Fallon J, Bunney WE (1997) Decreasing striatal 6-FDOPA uptake with increasing duration of cocaine withdrawal. Neuropsychopharmacology 17:402-409.

Xi ZX, Gilbert J, Campos AC, Kline N, Ashby CR, Jr., Hagan JJ, Heidbreder CA, Gardner EL (2004) Blockade of mesolimbic dopamine D3 receptors inhibits stress-induced reinstatement of cocaine-seeking in rats. Psychopharmacology (Berl) 176:57-65.

Yen SS, Quigley ME, Reid RL, Ropert JF, Cetel NS (1985) Neuroendocrinology of opioid peptides and their role in the control of gonadotropin and prolactin secretion. Am J Obstet Gynecol 152:485-493.

Young SN, Pihl RO, Benkelfat C, Palmour R, Ellenbogen M, Lemarquand D (1996) The effect of low brain serotonin on mood and aggression in humans. Influence of baseline mood and genetic factors. Adv Exp Med Biol 398:45-50.

Zakharova E, Leoni G, Kichko I, Izenwasser S (2009) Differential effects of methamphetamine and cocaine on conditioned place preference and locomotor activity in adult and adolescent male rats. Behav Brain Res 198:45-50.

Zaslav MR (1994) Psychology of comorbid posttraumatic stress disorder and substance abuse: lessons from combat veterans. J Psychoactive Drugs 26:393-400.

Zecevic N, Bourgeois JP, Rakic P (1989) Changes in synaptic density in motor cortex of rhesus monkey during fetal and postnatal life. Brain Res Dev Brain Res 50:11-32.

Zeleznik AJ (1981) Premature elevation of systemic estradiol reduces serum levels of follicle-stimulating hormone and lengthens the follicular phase of the menstrual cycle in rhesus monkeys. Endocrinology 109:352-355.

Zhou Y, Spangler R, Schlussman S, Ho A, Kreek M (2003) Alterations in hypothalamic-pituitary-adrenal axis activity and in levels of proopiomelanocortin and corticotropin-releasing hormone-receptor 1 mRNAs in the pituitary and hypothalamus of the rat during chronic 'binge' cocaine and withdrawal. Brain Res 964:187-199.

Zhou Y, Spangler R, LaForge KS, Maggos CE, Ho A, Kreek MJ (1996) Corticotropin-releasing factor and type 1 corticotropin-releasing factor receptor messenger RNAs in rat brain and pituitary during "binge"-pattern cocaine administration and chronic withdrawal. J Pharmacol Exp Ther 279:351-358.

Zito KA, Vickers G, Roberts DC (1985) Disruption of cocaine and heroin self-administration following kainic acid lesions of the nucleus accumbens. Pharmacol Biochem Behav 23:1029-1036.

65

Chapter 2

Methods

2.1. Animal Model

2.1.1. Introduction

The selection of an appropriate animal model is crucial to obtain conclusive data

out of the experimental design (Vorhees, 1995). As discussed in the first chapter the

effects of cocaine exposure have been studied in a variety of animal models.

Clinical studies exhibit a series of limitations in what concerns to the assessment

of drug abuse effects. In human studies it is difficult to exclude the influence of other

factors such as the use of other illegal and legal drugs, co-morbid psychiatric conditions,

and differences in lifestyle (Porrino et al., 2007). Because of these and many other

problems, it is virtually impossible to isolate and address the issue of the consequences of

chronic exposure in the human population (Porrino et al., 2007). One approach to

addressing this question is the use of animal models. Animal models enable to predict the

effects of drug exposure in humans by systematically manipulating variables and

controlling for many of the problems encountered in human studies (Porrino et al., 2007).

The usefulness of animal models depends in large part on their validity as

simulations of human psychopathology or the human behaviour (Willner, 1997). In the

case of drugs abuse, like humans, other animals voluntarily self-administer drugs, and

their reinforcing properties have been demonstrated in animals such as rodents and non-

human primates (Mello et al., 1997; Tidey and Miczek, 1997; Carroll et al., 2002).

Moreover, in general, there is close agreement between the drugs self-administered by

animals and those which are abuse by people (Willner, 1997). The possibility of studying

these behaviours in animals has helped to understand the neurobiological basis of drug

taking (Koob and Moal, 1997; Nestler and Aghajanian, 1997; Everitt and Wolf, 2002) and,

more generally, the brain systems for reward (Wise, 2002).

In the present study, the effects of chronic cocaine exposure during adolescence

period were analyzed. This evaluation was achieved using an animal model of exposure

to cocaine during adolescence in Wistar rats.

The developmental stage of adolescence is not uniquely human. Developing

organisms from other species likewise undergo adolescent-typical transitions that include

pubertal changes and a growth-spurt, along with expression of certain adolescent-typical

behavioural patterns. For instance, even when considering a simple animal model of

adolescence in the rat, animals undergoing this transition exhibit more risk taking, novelty-

seeking, and peer-directed social interactions than adults (Douglas et al., 2003, 2004;

Stansfield and Kirstein, 2006). In fact, similarities across species in adolescent-typical

physiological and behavioural characteristics, including a marked remodelation of the

69

brain, are consistent with the notion that adolescence is a highly conserved

developmental stage during evolution, with a number of specific adolescent typical

behaviour patterns postulated to confer adaptive significance (Spear, 2007). Across-

species similarities in physiological and behavioural attributes of adolescence provide

reasonable face and construct validity for the use of animal models to study potential

neurotoxic effects during adolescence (Spear, 2007).

A critical issue when assessing consequences of drug exposure during

adolescence is the timing of the exposure period (Spear, 2007). Adolescence lacks clear

markers of onset and offset (McCormick and Mathews, 2007). In rats, the age range from

28 to 42 days postnatal has been conservatively classified as adolescence, based on

timing of age-specific behavioural changes, neural changes in brain, puberty, and the

growth spurt (Spear, 2007). Yet, some harbingers of adolescence may begin in females

as early as postnatal day (PND) 23 or so, with some residual signs perhaps lasting until

55 days or so in males (Spear, 2007). Use of this conservative age range is not meant to

imply, however, that animals slightly younger or older than this prototypic age range might

not also be undergoing adolescent transitions (Spear, 2000). Thus, to ensure that an

exposure period blankets the entire adolescence period in rats of both sex, it might be

necessary to begin shortly after the conventional age of weaning and continue until at

least 55 days (Spear, 2007). Day 60 onward is commonly agreed upon as adulthood in

that the animal has achieved physical and sexual maturity (McCormick and Mathews,

2007).

For our animal model design we choose a more restricted exposure period. We

decided for a period that comprises the reproductive maturation and the pubertal

development, since organizational effects of gonadal steroids during puberty may be

particularly relevant to drug abuse problems (Laviola et al., 1999; Spear, 2000; Chambers

et al., 2003). Thus, we decided to coincide the beginning of the administration of cocaine

with the onset of the puberty. Physical markers have been used as estimates of puberty

onset (McCormick and Mathews, 2007). In male rats, a marker is balanopreputial

separation (separation of the prepuce from the glans penis) which occurs at approximately

40 days of age (Korenbrot et al., 1977). In females, a marker is vaginal opening which

occurs at approximately 35 days of age (Evans, 1986). However, there is a significant

variation in the literature for days of balanopreputial separation (32 to 46 days) and day of

vaginal opening (32 to 38 days) (McCormick and Mathews, 2007). Based on these data

the PND 35 was chosen for the beginning of the chronic administration of cocaine that

proceeded until PND 50. This period of exposure allows starting the evaluation of the

chronic exposure effects within the transition period to the adulthood.

70

When selecting an adequate animal model to assess the impact of a drug of

abuse, it is crucial to determine which dose, route of administration and frequency of

administration would be more satisfactory to the study in perspective (Spear, 1996). In the

design of our animal model was selected a binge pattern of cocaine administration,

consisting in three daily 15mg/kg intraperitoneal (i.p.) injections administered 1hour apart,

followed by no cocaine administration for 22 hours. The daily i.p. dose of 3x15mg/kg of

cocaine was selected based on previous studies in which this dose has was demonstrated

to induced neurobiological and behavioural alterations (Tsukada et al., 1996; Sarnyai et

al., 1998; Zhou et al., 2001; Schlussman et al., 2002; Zhou et al., 2003; Zhou et al., 2005;

Black et al., 2006). This is an elevated dose still, not enough to induce seizures

convulsion, or more several lethal consequences (Schlussman et al., 2005). The dosing

schedule was chosen to mimic the pattern of consumption often seen in human cocaine

addicts with respect to repeated administrations over a short time period, followed by a

period of abstinence (Gawin, 1991). For many drugs, a pattern of self-administration

develops under unlimited access conditions, consisting of periods of drug intake

alternating with periods of abstinence, during which other activities such as eating,

drinking and sleeping occur (Willner, 1997). This binge pattern of drug consumption has

been described in animals self-administering a wide variety of psychostimulants, and with

alcohol, and is also characteristic of the problematic use of both these drug classes in

humans (Willner, 1997).

2.1.2. Animals

Animals born from nulliparous three-month old female Wistar rats acquired from

Charles River Laboratories España S.A. (Barcelona, Spain) were used in this study.

Animals were housed in animal facility room of our laboratory at the Institute for Molecular

and Cell Biology, Porto, Portugal. Animals were maintained under stable conditions,

constant temperature (20-22 ºC) and humidity (60%) with a 12 hour light/dark cycle. Water

and appropriate food were supplied ad libitum. Institutional guidelines regarding animal

experimentation were followed. Rats were handled daily and regularly weighed.

Cylindrical plastic tubes and soft paper for nest construction were made available to

reduce stress. All procedures were approved by the Portuguese Agency for Animal

Welfare (General Board of Veterinary Medicine in compliance with the Institutional

Guidelines and the European Convention).

In the day after birth, here designated as postnatal day (PND) 1, litters were

standardized to 10 pups and balanced to 5 males and 5 females whenever possible.

71

Animals were weaned on PND 21 and on PND 30 animals housed individually in order to

avoid variable social influences that could play a role in cocaine behavioural,

neurochemical and hormonal effects (de Jong et al., 2007) and were randomly assigned

to the different experimental groups. Only one male and one female from each litter were

used in each experimental group. At least 6 animals were used in each experimental

group.

2.1.3 Chronic administration and assessment time points

Rats were treated following a binge pattern administration of cocaine. Animals

were daily injected 3 times i.p. with 15mg/kg of body weight cocaine hydrochloride, in

0.9% NaCl solution from PND 35 to PND 50. The daily injections were administered at

hourly intervals, between 9:00 and 11:00 a.m., in a volume of 1mL/kg. Controls were

given isovolumetric 0.9% NaCl solution following the same experimental protocol. Cocaine

hydrochloride was obtained from Sigma Chemicals (St. Louis, MO, USA) and prepared in

0.9% NaCl solution. During the entire period of drug administration animals were daily

weighed.

Figure 2.1.3.1- Schematic diagram representing the cocaine exposure period and the assessment time points

estasblished to evaluate the effects of cocaine administration throughout adolescence. PND- postnatal day.

The assessments were done at different time points after the end of the exposure period:

- at PND 50, 30 minutes after last binge cocaine administration of cocaine. This time point

of assessment allowed the study of the hormonal and neurochemical effects of the acute

binge cocaine administration during a chronic exposure;

- at PND 51 and 52, 1 and 2 days, after the end of the exposure period, respectively.

These time points of assessment allowed to obtain hormonal, neurochemical and

72

behavioural data of the short-term effects of the chronic cocaine administration during

adolescence period;

- at PND 55, 5 days after the end of the exposure period. This time point of assessment

allowed to evaluate, within the adolescence period, hormonal, neurochemical and

behavioural long-term effects of the chronic cocaine administration during adolescence;

- at PND 60, 10 days after the end of the exposure period. This time point of assessment

allowed to evaluate, in the early adulthood, the long-term effects of cocaine exposure

during the adolescence period, measured as neurochemical, hormonal, behavioural and

mRNA expression data.

2.2. Blood and tissue collection

At the end of the experimental periods animals were decapitated and brains were

rapidly removed. Brain areas related with the response to cocaine, such as substantia

nigra and ventral tegmental area (SN-VTA), Nac, dorsal striatum, prefrontal cortex,

amygdala, hippocampus, dorsal raphe nucleus, cerebellum and a hypothalamus section

that include suprachiasmatic, pre-optic and paraventricular nuclei were dissected on ice.

Dissection was processed according to the atlas by Paxinos and Watson (1998). Pituitary

glands were also collected. Tissue samples were frozen by immersion in 2-methylbutane

cooled over dry ice and stored at -70ºC until used for further analyses. Trunk blood was

collected in lithium heparin cover tubes and centrifuged at 1600g for 15 minutes, obtained

plasma was kept at –70ºC until hormonal assays. Gonads and adrenal glands were also

collected and weighed, and the data recorded.

2.3. Hormone plasma levels measures

All hormone plasma levels were measured by enzyme immunoassay, using

commercially available assays.

2.3.1. Steroid plasma levels

The progesterone, estradiol, testosterone and corticosterone plasma levels were

measured by enzyme immunoassay, using commercially available kit provided by

Cayman Chemical Company (Ann Arbor, MI, USA).

For progesterone, assay sensitivity was 0.012ng/mL, and the intra- and inter-assay

coefficients of variation were less 8% and 12%, respectively. For estradiol, assay

sensitivity was 0.004ng/mL, and the intra- and inter-assay coefficients of variation were

73

less 10% and 12%, respectively. For testosterone, assay sensitivity was 0.004ng/mL, and

the intra- and inter-assay coefficients of variation were less 10% and 15%, respectively.

For corticosterone assay sensitivity was 0.04ng/mL, and the intra- and inter-assay

coefficients of variation were less than 7% and 9%, respectively.

These assays are based on the competition between the sterois and an steroid-

acetylcholinesterase (AChE) conjugate for a limited number of steroid-specific rabbit

antiserum binding sites. Because the concentration of the steroid AChE conjugate is held

constant while the concentration of steroid varies, the amount of steroid AChE conjugate

that is able to bind to the rabit antiserum will be inversely proportional to the concentration

of the steroid present. A rabbit antiserum-steroid (either free or AChE conjugate) complex

binds to the mouse monoclonal anti-rabbit antibody. After removing the unbound reagent,

a colour reaction is developed using a substrate to the AChE. The intensity of this color,

determined spectrophotometrical, is proportional to the amount of the steroid AChE

conjugate, which is inversely proportional to the amount of free steroid present. Sample

concentrations were read from a calibration curve.

Corticosterone plasma levels after behavioural tests were measured by enzyme

immunoassay, using commercially available kit provided by Immunodiagnostic Systems

Ltd (Boldon, UK). Assay sensitivity was 0.23 ng/mL. The intra- and inter-assay coefficients

of variation were less than 3.1% and 7.2%, respectively. This assay is also a competitive

enzyme immunoassay. Samples are incubated with enzyme (horseradish peroxidase)

labelled corticosterone in the presence of the corticosterone antibody. The corticosterone

present in the samples competes with the horseradish peroxidase labelled corticosterone

to the corticosterone antibody. After removing the unbound reagent, a colour reaction is

developed using a chromogenic substrate to the horseradish peroxidase.The intensity of

this colour, determined spectrophotometrical, is inversely proportional to the concentration

of corticosterone. Sample concentrations were read from a calibration curve.

2.3.2. ACTH plasma levels

ACTH plasma levels were measured by enzyme immunoassay, using a

commercially available kit provided by Phoenix Pharmaceuticals, Inc. (Belmont, CA,

USA). For the ACTH assay the sensitivity was 0.18 ng/mL, and the intra- and inter-assay

coefficients of variation were less than 5 % and 14%, respectively.

The assay is based on the presence of a secondary antibody that can bind to the

fragment cristallizable (Fc) region of the primary antibody whose fragment, antigen

binding (Fab) region will be competitively bound by both biotinylated peptide or targeted

74

peptide in samples. The biotinylated peptide is able to interact with streptavidin-

horseradish peroxidase (SA-HRP) which catalyzes a substrate solution. The intensity of

the colour developed, determined spectrophotometrical, is directly proportional to the

amount of biotinylated peptide-SA-HRP complex but inversely proportional to the amount

of the peptide in samples. Sample concentrations were read from a calibration curve.

2.4. Neurotransmitters determinations

Levels of DA and 5-HT and their metabolites, 3,4-dihydroxyphenylacetic acid

(DOPAC), homovanillic acid (HVA) and 5-hydroxyindole-3-acetic acid (5-HIAA) were

quantified by a modified method of high-performance liquid chromatography with

electrochemical detection (HPLC-EC) (Ali et al., 1993), using a Gilson Medical Electronics

system (Gilson Inc., Middleton, WI, USA). The HPLC-EC system was composed of a

LC307 delivery pump, LC142 electrochemical detector, LC234 auto-injector, and 712

HPLC controller software version 1.30. The analytic column was a Supelcosil LC-18 3μM

(Supelco Inc., Bellefonte, PA, USA).

Prior to neurochemical determinations, 200 µl of 0.2 N perchloric acid were added

to the dissected brain regions. Samples were then disrupted by ultrasonication and

centrifuged (15,000 g) at 4ºC, during 5 minutes. Supernatant was collected and filtered

through a Costar micro centrifuge filter (0.2 µm nylon filter) (Corning Incorporated,

Corning, NY, USA). Aliquots of 50 µl were injected directly onto the HPLC-EC system for

the quantification of DA, 5-HT and their metabolites.

The mobile phase solution was composed of 70 mM potassium phosphate

monobasic buffer (pH was adjusted to 3.0 by adding with phosphoric acid), 10% (v/v)

methanol, 1mM 1-Heptanesulfonic acid and 107.5 μM sodium ethylenediaminetetraacetic

acid. The mobile phase was filtered through a 0.2 μm filter, degassed, and delivered at a

flow rate of 1.0 mL/min.

Concentrations of DA, 5-HT, DOPAC, HVA and 5-HIAA were calculated using

standard curves generated using 5 known amounts of standard amines purchased from

Sigma Chemicals (ST. Louis, MO, USA). Concentrations are expressed in ng/mg protein.

The pellet of each sample was ressuspended in 0.2 M phosphate buffer and, after

sonication, the total content of protein was assayed in duplicate using a colorimetric assay

from Bio-Rad (Munchen, Germany) based on the Bradford assay (Bradford, 1976).

Bovine serum albumin was used as a standard protein.

75

The ratios of DOPAC to DA, HVA to DA and DOPAC and HVA to DA were

determined for each animal and used as indexes of DA turnover rate, whereas the ratio of

5-HIAA to 5-HT was used as an index of 5-HT turnover rate.

2.5. mRNA expression levels determination

Total RNA was extracted from brain regions related with cocaine’s response using

the RNeasy® Lipid Tissue Mini kit (Qiagen, Austin, Texas, USA) and following the

instructions of the manufacturer. Purity was estimated from the ratio of absorbance

readings at 260 and 280 nm and only ratio between 1.8 e 2 were accepted. RNA quality

was checked in agarose gel. RNA concentration was determined in a NanoDrop

spectrophotometer (Thermo Scientific, Wilmington, Delaware, USA). One µg of RNA was

reverse transcribed, using the SuperScriptTM First-Strand Synthesis System for RT-PCR

(Invitrogen, Carlsbad, CA, USA), using oligo(dt) primers and following the instructions of

the manufacturer.

The expression levels of mRNA transcripts for the different genes studied were

measured by quantitative real-time polymerase chain reaction (qRT-PCR). The reference

gene, glyceraldahyde-3-phosphate-dehydrogenase (GAPDH) was used as internal

standard for normalization. The qRT-PCR reactions, using the equal amounts of total RNA

from each sample, were performed on the iQ5 Multicolor Real-time PCR Detection

System from Bio-Rad (Hercules, CA, USA), using the iQTM SYBR® Green Supermix (Bio-

Rad Laboratories, Hercules, CA, USA). Product fluorescence was detected at the end of

the elongation cycle. All melting curves exhibited a single sharp peak at a temperature

characteristic of the primer used. Primer design was performed using the software Beacon

Designer (Premier Biosoft International, Palo Alto, CA, USA).

Linearity and efficiency of PCR amplification reactions were assessed for all pair of

primers using standard curves generated by increasing amounts of cDNA. The

relationship between the threshold cycle (Ct) and logarithm of the cDNA concentrations

were studied according to correlation coefficient and the slopes, calculated by Bio-Rad

iQ5 Optical System Software, version 2.0 (Bio-Rad Laboratories, Inc, 2006). For all primer

sets, standard curves using four points, diluted over a 100-fold range, always led to a high

linearity (correlation coefficients>I- 0.990I).

The PCR efficiency (Ex) was calculated using the equation Ex= 10-1/slope. Efficiency

was presented as a percentage of the template that was amplified in each cycle,

calculated by the following equation %Ex= (EX-1) x 100. Efficiency close to 100% is the

best indicator of a robust, reproducible assay. Efficiency between 90-105% was always

76

used. These amplification efficiencies of PCR assays allow the quantification of mRNA

with the comparative Ct quantification method (ΔCt method) using a reference gene.

Following this method, the relative expression of a gene was calculated by the expression:

2 (Ct(referencia) – Ct(target) ). This method assumes that both target and reference genes are

amplified with efficiencies near 100% and within 5% of each other.

2.6. Behavioural tests

2.6.1. Elevated plus maze test

The anxiety-like behaviour was assessed using the elevated plus maze test

(EPM), which exploits the conflict between the animal's innate tendency to explore novel

areas and their aversion for heights and open spaces (Montgomery, 1955; Handley and

Mithani, 1984).

The EPM apparatus consisted of four arms 46 cm long and 14 cm wide with two

arms open and two arms with 22 cm high walls. The maze was mounted 74 cm above the

floor under fluorescent lighting (300 lux).

At the time of the test, rats were placed individually on the central portion of the

EPM apparatus, facing an open arm. They were allowed to explore the apparatus for 5

minutes and were recorded using a video camera placed 1m above the apparatus.

Videotapes were analysed using the Noldus Observer software (Observer 4.1;

Noldus Information Technology, Wageningen, Netherlands). Spatiotemporal and

behavioural parameters were recorded. Spatiotemporal measures comprised the

frequencies of total, open and closed arm entries; central platform entries; the percentage

of open-arm entries (open/total x 100); and the time spent in the open, closed or central

parts of the maze. Behavioural measures comprised frequency and duration scores for

rearing (vertical movements, animal sustained in posterior paws with anterior paws

against or not to the walls), head dipping (exploratory movement of head and shoulders

over the edge of the maze) and grooming (licking the paws, washing movements over the

head, fur licking, and tail and genitals cleaning).

2.6.2 Forced swim test

The forced swim test (FST) is a behavioural paradigm that is widely used as a

predictor of antidepressant activity in rodents (Porsolt et al., 1977). In this paradigm rats

are forced to swim in water for 15 minutes on the first day of testing, and for 5 minutes on

77

the second day of testing. On the second day of testing behavioural monitoring reveals

that the animal quickly adopts an immobile posture, and this is said to reflect a state of

“behavioural despair”. Subchronic and chronic antidepressant treatment largely attenuates

FSH induced immobility (Borsini and Meli, 1988).

The FST procedure used was similar to the one described by Porsolt et al. (1977).

At the time of test rats were placed into a glass cylinder (19 cm diameter, 40 cm deep) of

22±1ºC water filled to a depth of 30 cm for 15 min of pre-test, under fluorescent lighting.

Twenty-four hours later rats were again placed into the water for 5 min. Activity during pre-

test and second swim test was video-recorded for subsequent behavioural analysis.

Videotapes analysed using the Noldus Observer software (Observer 4.1 Noldus

Information Technology, Wageningen, Netherlands) and the following behaviour

categories were analyzed: immobility, struggling, diving, as well as right and left rotation.

The data was collected for frequency, duration and latency. Immobility was defined as

lack of movement of three paws or lack of movement of all four paws. Struggling was

defined as vigorous movement of all four paws, with the rat in a relatively upright position.

Diving was defined as under water swim.

2.6.3. Aggressiveness test

The aggressiveness test was conducted during the dark phase of the diurnal cycle,

utilizing the procedures from the resident-intruder paradigm as described by (Miczek,

1979). The resident-intruder paradigm is used to evaluate the aggressive behaviour of

the resident rat face to an intruder rat, that strongly resembles the natural patterns of wild

rat to establish and defend their territory (Koolhaas et al., 1980).

The experimental animal was always the resident and the intruder was an

unfamiliar, non-experimental age-matched male rat and with similar weight. The resident

rat, in its home cage, was allowed to habituate to the room test for 40 minutes, following

which an unfamiliar intruder was placed in its home cage and their behaviour was

videotaped for 10 minutes. The recording area was illuminated by red light illumination.

Behaviour of the resident was recorded and later analysed. Frequency, duration and

latency data were collected for the following behaviours divided in to three categories: (1)

offensive behaviours that included attack, bite, pinning, wrestling, chasing and aggressive

grooming; (2) defensive behaviours that included flight, avoidance of contact and

submissive-supine posture; (3) social investigation behaviours including sniffing,

anogenital sniffing, and approach behaviour. Attack was considered when the

experimental subject lunges at the partner with its forepaws extended outward; pinning

78

was considered when the resident subject stands over the exposed ventral area of the

partner pressing it against the floor; wrestling was considered when the two animals roll

and tumble with one another; chasing was considered when the experimental animal

rapidly pursues the intruder; aggressive grooming was consider when vigorous grooming

of the immobile partner using the teeth and the forepaws; flight was considered when the

experimental animal were pursued by the intruder; supine posture was considered when

the intruder stands over the exposed ventral areas of the resident animals pressing it

against the floor; avoidance of contact was considered when the resident move away from

the intruder; anogenital sniffing was considered when the resident sniffs the anogenital

region of the intruder; approach behaviour was considered when the resident move in to

the intruder direction.

2.7. Statistical analysis

Data were analyzed by appropriate statistical test (reported in more detail in the

following chapters). Statistical analyses were performed using the software SPSS 14.0

(SPSS INC. Chicago, Illinois, USA). Differences were considered to be statistically

significant when p<0.05. All data are expressed as the mean and standard error of the

mean (S.E.M.).

79

2.8. References

Ali SF, David SN, Newport GD (1993) Age-related susceptibility to MPTP-induced neurotoxicity in mice. Neurotoxicology 14:29-34.

Black YD, Maclaren FR, Naydenov AV, Carlezon WA, Jr., Baxter MG, Konradi C (2006) Altered attention and prefrontal cortex gene expression in rats after binge-like exposure to cocaine during adolescence. J Neurosci 26:9656-9665.

Borsini F, Meli A (1988) Is the forced swimming test a suitable model for revealing antidepressant activity? Psychopharmacology (Berl) 94:147-160.

Bradford MM (1976) A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 72:248-254.

Carroll ME, Morgan AD, Lynch WJ, Campbell UC, Dess NK (2002) Intravenous cocaine and heroin self-administration in rats selectively bred for differential saccharin intake: phenotype and sex differences. Psychopharmacology (Berl) 161:304-313.

Chambers RA, Taylor JR, Potenza MN (2003) Developmental neurocircuitry of motivation in adolescence: A critical period of addiction vulnerability. Am J Psychiatry 160:1041-1052.

de Jong IE, Oitzl MS, de Kloet ER (2007) Adrenalectomy prevents behavioural sensitisation of mice to cocaine in a genotype-dependent manner. Behav Brain Res 177:329-339.

Douglas LA, Varlinskaya EI, Spear LP (2003) Novel-object place conditioning in adolescent and adult male and female rats: effects of social isolation. Physiol Behav 80:317-325.

Douglas LA, Varlinskaya EI, Spear LP (2004) Rewarding properties of social interactions in adolescent and adult male and female rats: impact of social versus isolate housing of subjects and partners. Dev Psychobiol 45:153-162.

Evans AM (1986) Age at puberty and first litter size in early and late paired rats. Biol Reprod 34:322-326.

Everitt BJ, Wolf ME (2002) Psychomotor stimulant addiction: a neural systems perspective. J Neurosci 22:3312-3320.

Gawin FH (1991) Cocaine addiction: psychology and neurophysiology. Science 251:1580-1586.

Handley SL, Mithani S (1984) Effects of alpha-adrenoceptor agonists and antagonists in a maze-exploration model of 'fear'-motivated behaviour. Naunyn Schmiedebergs Arch Pharmacol 327:1-5.

Koob GF, Moal ML (1997) Drug abuse: hedonic homeostatic dysregulation. Science 278:52-58.

Koolhaas JM, Schuurman T, Wiepkema PR (1980) The organization of intraspecific agonistic behaviour in the rat. Prog Neurobiol 15:247-268.

Korenbrot CC, Huhtaniemi IT, Weiner RI (1977) Preputial separation as an external sign of pubertal development in the male rat. Biol Reprod 17:298-303.

Laviola G, Adriani W, Terranova ML, Gerra G (1999) Psychobiological risk factors for vulnerability to psychostimulants in human adolescents and animal models. Neuroscience and Biobehavioral Reviews 23:993-1010.

McCormick CM, Mathews IZ (2007) HPA function in adolescence: role of sex hormones in its regulation and the enduring consequences of exposure to stressors. Pharmacol Biochem Behav 86:220-233.

Mello NK, Mendelson JH, Kelly M, Diaz-Migoyo N, Sholar JW (1997) The Effects of Chronic Cocaine Self-Administration on the Menstrual Cycle in Rhesus Monkeys. J Pharmacol Exp Ther 281:70-83.

Miczek KA (1979) A new test for aggression in rats without aversive stimulation: differential effects of d-amphetamine and cocaine. Psychopharmacology (Berl) 60:253-259.

Montgomery KC (1955) The relation between fear induced by novel stimulation and exploratory behavior. J Comp Physiol Psychol 48:254-260.

80

Nestler EJ, Aghajanian GK (1997) Molecular and cellular basis of addiction. Science 278:58-63.

Paxinos G, Watson C (1998) The rat brain in stereotaxic coordinates, Fourth Edition. Academic Press, San Diego.

Porrino LJ, Smith HR, Nader MA, Beveridge TJ (2007) The effects of cocaine: a shifting target over the course of addiction. Prog Neuropsychopharmacol Biol Psychiatry 31:1593-1600.

Porsolt RD, Le Pichon M, Jalfre M (1977) Depression: a new animal model sensitive to antidepressant treatments. Nature 266:730-732.

Sarnyai Z, Dhabhar FS, McEwen BS, Kreek MJ (1998) Neuroendocrine-related effects of long-term, 'binge' cocaine administration: Diminished individual differences in stress-induced corticosterone response. Neuroendocrinology 68:334-344.

Schlussman S, Nyberg F, Kreek M (2002) The effects of drug abuse on the stress responsive hypothalamic-pituitary-adrenal axis and the dopaminergic and endogenous opioid systems. Acta Psychiatr Scand Suppl:121-124.

Schlussman S, Zhou Y, Bailey A, Ho A, Kreek M (2005) Steady-dose and escalating-dose "binge" administration of cocaine alter expression of behavioral stereotypy and striatal preprodynorphin mRNA levels in rats. Brain Research Bulletin 67:169-175.

Spear L (2000) The adolescent brain and age-related behavioral manifestations. Neurosci Biobehav Rev 24:417-463.

Spear LP (1996) Assessment of the effects of developmental toxicants: pharmacological and stress vulnerability of offspring. NIDA Res Monogr 164:125-145.

Spear LP (2007) Assessment of adolescent neurotoxicity: rationale and methodological considerations. Neurotoxicol Teratol 29:1-9.

Stansfield KH, Kirstein CL (2006) Effects of novelty on behavior in the adolescent and adult rat. Dev Psychobiol 48:10-15.

Tidey JW, Miczek KA (1997) Acquisition of cocaine self-administration after social stress: role of accumbens dopamine. Psychopharmacology (Berl) 130:203-212.

Tsukada H, Kreuter J, Maggos CE, Unterwald EM, Kakiuchi T, Nishiyama S, Futatsubashi M, Kreek MJ (1996) Effects of binge pattern cocaine administration on dopamine D1 and D2 receptors in the rat brain: An in vivo study using positron emission tomography. J Neurosci 16:7670-7677.

Vorhees CV (1995) A review of developmental exposure models for CNS stimulants: cocaine In: Mothers, babies and cocaine: the role of toxins in development (Lewis, Bendersky, eds), pp 71-94. Hillsdale: Lawrence Eribaum Associates.

Willner P (1997) Animal models of addiction. Human Psychopharmacology-Clinical and Experimental 12:S59-S68.

Wise RA (2002) Brain reward circuitry: insights from unsensed incentives. Neuron 36:229-240.

Zhou Y, Spangler R, Ho A, Jeanne Kreek M (2001) Hypothalamic CRH mRNA levels are differentially modulated by repeated 'binge' cocaine with or without D(1) dopamine receptor blockade. Brain Res Mol Brain Res 94:112-118.

Zhou Y, Spangler R, Schlussman S, Ho A, Kreek M (2003) Alterations in hypothalamic-pituitary-adrenal axis activity and in levels of proopiomelanocortin and corticotropin-releasing hormone-receptor 1 mRNAs in the pituitary and hypothalamus of the rat during chronic 'binge' cocaine and withdrawal. Brain Res 964:187-199.

Zhou Y, Bendor J, Yuferov V, Schlussman S, Ho A, Kreek M (2005) Amygdalar vasopressin mRNA increases in acute cocaine withdrawal: Evidence for opioid receptor modulation. Neuroscience 134:1391-1397.

81

Chapter 3

Short-term Effects of Cocaine Exposure throughout Adolescence in the Rat

3.1. Introduction

Although adolescents represent a risk group regarding drug abuse and addiction,

experimental research evaluating the effects of psychostimulants has been mostly

focused on the assessment of adult animals. In fact, previous studies have shown that

most elicit drug use begins in adolescence (Estroff et al., 1989; Chen and Kandel, 1995),

that adolescents are more likely to progress from use to dependence than adults (Chen et

al., 1997) and do so much faster (Clark et al., 1998).

Adolescence is an ontogenetic transition to adulthood during which the brain and

hormonal systems are still undergoing many complex changes (Spear, 2000). Disruption

of the development occurring at this stage may increase the probability of developing

psychopathologies such as drug addiction (reviewed by Spear, 2000).Therefore, there is a

growing interest in assessing the influence that drug use can have on the developing

adolescent.

It have been reported that adolescents differ from adults in their hormonal,

neurochemical and behavioural response to psychostimulants (e.g., Adriani and Laviola,

2000; Collins and Izenwasser, 2002).

Based on evidence from behavioural models of drug abuse and addiction, such as

locomotor sensitization, conditioned place preference, and self-administration,

adolescents appear to be more vulnerable to the rewarding properties and detrimental

effects on the nervous system of various drugs (see reviews by Tirelli et al., 2003; Leslie

et al., 2004; Barron et al., 2005).

The work described in this chapter was performed to evaluate the short-term

effects of chronic cocaine exposure during the adolescence period upon the activity of the

HPA and the HPG axes of both male and female Wistar rats, by assessing the hormonal

plasma levels 30 minutes and 1 day after the last administration of cocaine. The effects of

cocaine exposure were also evaluated upon the dopaminergic and serotonergic systems

by assessing the monoamines levels in brain regions known to be involved in the drug

addiction processes.

It was shown that cocaine abuse and withdrawal is linked to the development of

depressive and/or anxiety-like symptoms (Gawin, 1991; Markou et al., 1992). Moreover,

severe anxiety and depression provide part of the negative reinforcement associated with

cocaine dependence and are important motivational factors for relapse and maintenance

of repetitive cycles of cocaine abuse (Gawin et al., 1989; Markou et al., 1992; Goeders et

al., 1993; Shaham et al., 2000; Shalev et al., 2002).

85

Therefore, it was also our goal to evaluate the short-term effects of chronic cocaine

exposure during adolescence period on the behavioural strategies adopted to cope with

an environmental anxiogenic situation, by submitting the animals, 2 days after the last

administration, to the EPM. The presence of depressive-like symptoms were also verified

3 days after withdrawal from chronic cocaine exposure during adolescence, by assessing

the behaviour of both male and female adolescent rats in a FST. The effects of cocaine

treatment on the subsequent alterations on brain neurotransmitters and in the HPA axis

activity induced by the EPM or the FST were analysed.

86

3.2. Material and Methods

3.2.1. Animals

Using the animal paradigm described in Chapter 2, we have obtained animals

chronically exposed to cocaine during adolescence that were used in the following

experiments. Briefly, on PND 30 animals were housed individually, and were randomly

assigned to the different experimental groups. Only one male and one female from each

litter were used in each experimental group. Rats were treated following a binge pattern

administration of cocaine, each day being daily injected 3 times intraperitonealy with

15mg/kg of body weight cocaine hydrochloride dissolved in 0.9% NaCl solution from PND

35 to PND 50. The daily injections were administered at hourly intervals, between 9:00

and 11:00 a.m., in a volume of 1 mL/kg. Controls were given an isovolumetric 0.9% NaCl

solution following the same experimental protocol. During the entire period of drug

administration animals were daily weighed. At least 6 animals were used in each

experimental group.

3.2.2. Behavioural tests

3.2.2.1. Elevated plus maze test

On PND 52, 2 days after cocaine withdrawal, both male and female Wistar rats

were tested in an EPM apparatus as described in Chapter 2. Spatiotemporal and

behavioural parameters were registered at the time of behaviour analyses. Spatiotemporal

measures comprised the frequencies of total, open and closed arm entries and central

platform entries, as well as the time spent in the open, closed and central part of the

maze. Behavioural measures comprised frequency and duration scores for rearing and

head dipping and grooming.

3.2.2.2. Forced swim test

Starting on PND 52, 2 days after withdrawal, both male and female Wistar rats

performed the FST as described in Chapter 2. Behavioural activity was analysed and data

collected for frequency, duration and latency of immobility, struggling and diving, and

frequency and latency of right and left rotation.

87

3.2.3. Blood and tissue collection

Thirty minutes and 1 day after the last administration of cocaine, and after the

behavioural tests, animals were sacrificed by decapitation and trunk blood was collected

in lithium heparin cover tubes and centrifuged at 1600g for 15 minutes; the obtained

plasma was kept at -70ºC until hormonal assays were conducted. Brains were also rapidly

removed and SN-VTA, Nac, dorsal striatum, prefrontal cortex, amygdala, hippocampus,

dorsal raphe nucleus, cerebellum and a hypothalamic section containing the

suprachiasmatic, pre-optic and paraventricular nuclei were dissected on ice. Tissue

samples were frozen by immersion in 2-methylbutane cooled over dry ice and stored at -

70ºC until used for further analyses. Thirty minutes and 1 day after the last administration

of cocaine gonads and adrenal glands were collected and weighed, and the data obtained

was recorded.

3.2.4. Hormone plasma levels determinations

The levels of progesterone, estradiol, testosterone, corticosterone, and ACTH

were measured on plasma obtained 30 minutes and 1 day after the last administration.

The levels of corticosterone were also measured on the plasma obtained after the

behavioural tests. All hormones plasma levels were measured by enzyme immunoassay,

using commercially available assays.

The progesterone, estradiol, testosterone and corticosterone basal plasma levels

were measured using assays provided by Cayman Chemical Company (Ann Arbor, MI,

USA). For progesterone, the assay sensitivity was 0.012 ng/mL, and the intra- and inter-

assay coefficients of variation were less 8% and 12%, respectively. For estradiol, the

assay sensitivity was 0.004 ng/mL, and the intra- and inter-assay coefficients of variation

were less 10% and 12%, respectively. For testosterone, the assay sensitivity was 0.004

ng/mL, and the intra- and inter-assay coefficients of variation were less 10% and 15%,

respectively. For corticosterone, the assay sensitivity was 0.04 ng/mL, and the intra- and

inter-assay coefficients of variation were less than 7% and 9%, respectively. The ACTH

plasma levels were measured using an assay provided by Phoenix Pharmaceuticals, Inc.

(Belmont, California, USA). The assay sensitivity was 0.18 ng/mL, and the intra- and inter-

assay coefficients of variation were less than 5 % and 14%, respectively.

Corticosterone plasma levels after the behavioural tests were measured using a kit

provided by Immunodiagnostic Systems Ltd (Boldon, UK). Assay sensitivity was 0.23

ng/mL. The intra- and inter-assay coefficients of variation were less than 3.1% and 7.2%,

respectively.

88

3.2.5. Neurochemical determinations

Concentrations of DA and 5-HT and their metabolites, 5-HIAA, DOPAC, and HVA ,

30 min. and 1 day after last administration of cocaine, were quantified by HPLC-EC, in the

dissected brain regions (SN-VTA, Nac, dorsal striatum, prefrontal cortex, amygdala,

hippocampus, dorsal raphe nucleus, cerebellum and hypothalamus), as described in

Chapter 2. After each behavioural test, levels of DA and 5-HT and their metabolites were

determined in the SN-VTA, dorsal raphe nucleus, amygdala, hippocampus, prefrontal

cortex and hypothalamus. Concentrations are expressed in ng/mg protein. The ratios of

DOPAC to DA, HVA to DA and DOPAC and HVA to DA were determined for each animal

and used as indexes of DA turnover rate, whereas the ratio of 5-HIAA to 5-HT was used

as an index of 5-HT turnover rate.

3.2.6. Statistical analysis

A 2-way analysis of variance (ANOVA) with treatment (cocaine or saline) and sex

as between-subject fixed-factors with repeated measures was used to evaluate body

weight evolution during the experimental period until acute withdrawal. Student’s t-test

was used as a post-hoc test to analyse statistical significant differences.

Adrenal and gonadal weights were converted in percentage of body weight, and

adrenal data was analyzed by a 3-way ANOVA with time point of assessment (30 minutes

and 1 day), treatment and sex as between-subject fixed-factors. Gonadal data was

analyzed by a 2-way ANOVA (time point of assessment (30 minutes and 1 day) x

treatment), gender separately.

For the hormonal and neurochemical data obtained 30 minutes and 1 day after the

last administration of cocaine and after each behavioural test, statistical significance

between groups was assessed using the non-parametric Mann-Whitney U Test.

The EPM and FST behaviour data were analysed by a 2-way ANOVA with

treatment (cocaine or saline) and sex as between-subject fixed-factors and behavioural

measurements as the dependent variable. Student’s t-test was used as a post-hoc test to

analyse statistical significant differences. Whenever data did not pass normality and

homogeneity of variance tests the non-parametric Mann-Whitney U Test was used.

Differences were considered to be statistically significant when p<0.05. All data

were expressed as the mean plus S.E.M.. Statistical analyses were performed using the

software SPSS 14.0 (SPSS INC. Chicago, Illinois, USA).

89

3.3. Results

3.3.1. Analyses performed 30 minutes and 1 day after the last administration of cocaine

3.3.1.1. Biometric measures

Evolution of body weight. Cocaine-treated and control animals were weight

everyday from PND 35 to PND 52, and data was analyzed by a 2-way ANOVA (treatment

x sex) with repeated measures. Weight gain was statistically different between male and

female animals [F(1,37)= 155.627; p<0.001]), with post-hoc comparisons using Student’s

t-test showing that, except for PND 36, the daily weight gain was higher in males than in

females (figure 3.3.1.1.1). The statistical analysis revealed no significant effects of the

cocaine treatment on the weight of male and male.

0

30

60

90

120

150

35 37 39 41 43 45 47 49 51

PND

Wei

ght g

ain

(g)

saline male cocaine male saline female cocaine female

Figure 3.3.1.1.1- Effects of chronic cocaine exposure on body weight evolution during experimental perio until

acute withdrawal. Values are mean ± S.E.M.. Saline male, n=11; cocaine male, n=11; saline female, n=12;

cocaine female, n=8. PND- postnatal day. &, sex effect; p<0.001.

Adrenal weight. Adrenals were collected 30 minutes and 1 day after the last

administration of cocaine and their weight were recorded. The weight data was converted

in percentage of body weight and was analyzed by a 3-way ANOVA (time point of

assessment (30 minutes or 1 day) x treatment x sex). The statistical analysis revealed a

main effect of sex [F(1,37)= 67.195; p<0.001] in the adrenal weight (figure 3.3.1.1.2); with

female rats showing higher values than male ones. No treatment effects were found in

both male and female rats and the adrenal weight also did not differ 30 minutes and 1 day

after the last administration.

&

90

Gonadal weight. Testes and ovaries were collected 30 minutes and 1 day after

the last administration of cocaine and their weight were recorded. The percentage of body

weight was calculated and data was analyzed by a 2-way ANOVA (age x treatment). The

statistical analysis revealed no significant effects of the cocaine treatment on the weight of

male and female gonads, both when assessed 30 min or 1 day after the last

administration (figure 3.3.1.1.3). There were also no differences between time points of

assessment in both male and female gonadal weights.

Adrenal glands

0

0.02

0.04

0.06

30 min. 1 day

% o

f bod

y w

eigh

t

saline male cocaine male saline female cocaine female

Figure 3.3.1.1.2- Effects of chronic cocaine exposure throughout adolescence on adrenal weight of male and

female rats assessed 30 minutes and 1 day after the last administration. Each column represents the mean +

S.E.M. for a 6 animals per group.

A Male gonads B Female gonads

0

0.4

0.8

1.2

30 min. 1 day

% o

f bod

y w

eigh

t

saline male cocaine male

0

0.03

0.06

0.09

30 min. 1 day

% o

f bod

y w

eigh

t

saline female cocaine female 3.3.1.1.3- Effects of chronic cocaine exposure throughout adolescence on (A) male gonads and (B) females

gonads weights assessed 30 minutes and 1 day after the last administration of cocaine. Each column

represents the mean + S.E.M. for a 6 animals per group.

91

3.3.1.2. Hormones plasma levels

The levels of hormones were measured on plasma obtained 30 minutes and 1 day

after the last administration of cocaine. The data was analyzed by the non-parametric

Mann-Whitney U Test.

ACTH. Cocaine-treated males presented increased levels of ACTH 30 minutes

after the last administration, when compared with controls [Z=-2.402 (2-tailed) p<0.02]

(figure 3.3.1.2.1-A). This difference was no longer observed at 1 day after withdrawal.

Both 30 minutes and 1 day after the last administration there were no significant

differences in ACTH levels between cocaine-treated and control females. Between 30

minutes and 1 day after the last administration, control males presented an increase in

ACTH plasma levels [Z=-2.242 (2-tailed) p<0.05].

Corticosterone. No significant differences were found on the plasma levels of

corticosterone between the experimental groups (figure 3.1.2.1-B).

Testosterone. In the male group, 30 minutes after the last administration, cocaine-

treated males presented lower levels of testosterone [Z=-2.242 (2-tailed) p<0.05] (figure

3.3.1.2.2-A). This difference was no longer observed 1 day after withdrawal. In the female

group (figure 3.3.1.1.5-B), there were no differences in the levels of testosterone between

cocaine-treated and control females both at 30 minutes and 1 day after the last

administration. However, there was a decrease in testosterone levels in cocaine-treated

females between the two assessed time points [Z=-2.402 (2-tailed) p<0.02].

Progesterone. No significant differences were found on the plasma levels of

progesterone between cocaine-treated rats and their respective controls and between

assessment time points. However, a trend for increased progesterone levels was found in

cocaine-treated males 30 minutes after the last administration [Z=-1.922 (2-tailed)

p=0.055] (figure 3.3.1.2.2- C).

Estradiol. There were no significant differences in plasma levels of estradiol

between cocaine-treated and control females (figure 3.3.1.1.5-E) at the two assessed time

points, and there were also no differences in the plasma levels between 30 minutes and 1

day after the last administration, both in cocaine-treated and control animals.

92

A

0

2

4

6

8

30 min. 1 day

AC

TH le

vels

(ng/

mL)

saline male cocaine male saline female cocaine female

B

0

40

80

120

160

30 min. 1 day

Cor

ticos

tero

ne le

vels

(ng/

mL)

saline male cocaine male saline female cocaine female

Figure 3.3.1.2.1- Effects of chronic cocaine exposure throughout adolescence on (A) ACTH and (B)

corticosterone plasma levels assessed 30 minutes and 1 day after the last administration of cocaine. Each

column represents the mean + S.E.M. for a 6 animals per group. ACTH- Adrenocorticotropic hormone.

Columns with the same letter are significantly different from each other. a,b p<0.05.

a,b

b a

93

A B

0.0

0.6

1.2

1.8

30 min. 1 dayTest

oste

rone

leve

ls

(ng/

mL)

saline male cocaine male

0.0

0.1

0.2

0.3

30 min. 1 dayTest

oste

rone

leve

ls

(ng/

mL)

saline female cocaine female

C D

0.0

0.2

0.4

0.6

30 min. 1 dayProg

este

rone

leve

ls

(ng/

mL)

saline male cocaine male

0

50

100

150

30 min. 1 dayProg

este

rone

leve

ls

(ng/

mL)

saline female cocaine female

E

0

15

30

45

30 min. 1 day

Estra

diol

leve

ls

(pg/

mL)

saline female cocaine female

Figure 3.3.1.2.2- Effects of chronic cocaine exposure throughout adolescence on plasma levels of

testosterone in (A) males and (B) females, progesterone in (C) males and (D) females and estradiol in (E)

females, assessed 30 minutes and 1 day after the last administration of cocaine. Each column represents the

mean + S.E.M. for a 6 animals per group. Columns with the same letter are significantly different from each

other. a p<0.05, b p<0.02.

a

a

b

b

94

3.3.1.3. Neurochemical determinations

The levels of DA, 5-HT and their metabolites, DOPAC, HVA and 5-HIAA were

measured by HPLC-EC 30 minutes and 1 day after the end of the exposure period, in the

following brain regions: SN-VTA, Nac, dorsal striatum, prefrontal cortex, hippocampus,

amygdala, hypothalamus, dorsal raphe nucleus and cerebellum. These different brain

regions were selected based on their involvement in cocaine response. The data obtained

was analyzed by the non-parametric Mann-Whitney U Test.

Within the SN-VTA, 30 minutes after the last administration of cocaine, no

differences were found on the levels of DA, 5-HT and their metabolites between cocaine-

treated and control male rats, or between cocaine-treated and control female rats (figure

3.3.1.3.1). However, 30 minutes after the last administration, statistical analysis revealed

that DOPAC levels were higher in cocaine-treated males than in cocaine-treated females

[Z=-2.008 (2-tailed) p<0.05], and this effect was not observed in the control group.

Moreover, in cocaine exposed females the levels of DOPAC increased from 30 minutes to

1 day after the last administration of cocaine [Z=-2.082 (2-tailed) p<0.05]. At 1 day after

the end of the exposure period, the levels of DA, DOPAC and 5-HT in SN-VTA were

increased in cocaine-treated males as compared to control males (DA: [Z=-2.315 (2-

tailed) p<0.05]; DOPAC: [Z=-2.430 (2-tailed) p<0.02]; 5-HT: [Z=-2.083 (2-tailed) p<0.05]).

The DA turnover in the SN-VTA was analyzed, as given by the ratios of DOPAC+HVA to

DA and each one of its metabolites to DA (DOPAC/DA and HVA/DA), between cocaine-

treated and control males or females both 30 min and 1day after the end of the exposure

period and no statistical differences were found (table 3.3.1.3.1). However, in males the

DOPAC/DA ratio increased from 30 minutes to 1 day after the last administration in

cocaine-treated rats but not in controls [Z=-2.030 (2-tailed) p<0.05]. The 5-HT turnover

was assessed as the ratio of 5-HIAA to 5-HT and statistical analysis revealed no

differences between groups.

Within the Nac, no differences were detected in the levels of DA and 5-HT and

their metabolites between cocaine-treated rats and their respective controls at both time

points of analyses (figure 3.3.1.3.2). Also, no differences were found on both DA and 5-HT

turnover rates between the experimental groups (table 3.3.1.3.2).

95

SN-VTA

A

0

200400

600800

10001200

1400

DA DOPAC HVA DA DOPAC HVA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

30 min. 1 day

B

0

200

400

600

800

1000

1200

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

30 min. 1 day

Figure 3.3.1.3.1- Effects of chronic cocaine exposure throughout adolescence on the (A) levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the SN-VTA,

determined by HPLC-EC for male and female rats, 30 minutes and 1 day after the last administration. Each

column represents the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for

at least 5 animals per group. Columns with the same letter are significantly different from each other. a,b,c,d,e

p<0.05.

a a,d

b b

c c

e e

d

96

Nucleus accumbens

A

0

1500

3000

4500

6000

7500

DA DOPAC HVA DA DOPAC HVA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

30 min. 1 day

B

0

200

400

600

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

30 min. 1 day

Figure 3.3.1.3.2- Effects of chronic cocaine exposure throughout adolescence on the (A) levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the Nac, determined

by HPLC-EC for male and female rats, 30 minutes and 1 day after the last administration. Each column

represents the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for at least

6 animals per group.

97

Table 3.3.1.3.1- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the SN-VTA, 30 minutes and 1 day after the end of cocaine exposure period.

30 min. 1 day

Male Female Male Female

saline cocaine saline cocaine saline cocaine saline cocaine

DOPAC/DA 0.22±0.05

(n=6)

0.13±0.01

(n=5)

0.20±0.06

(n=6)

0.25±0.12

(n=6)

0.16±0.02

(n=8)

0.17±0.02

(n=7)

0.22±0.08

(n=6)

0.21±0.07

(n=6)

HVA/DA 0.06±0.02

(n=6)

0.06±0.01

(n=5)

0.14±0.10

(n=6)

0.15±0.09

(n=6)

0.08±0.02

(n=8)

0.07±0.02

(n=7)

0.05±0.01

(n=6)

0.06±0.01

(n=6)

(DOPAC+HVA)/DA 0.28±0.05

(n=6)

0.19±0.01

(n=5)

0.34±0.15

(n=6)

0.40±0.21

(n=6)

0.24±0.02

(n=8)

0.24±0.02

(n=7)

0.27±0.08

(n=6)

0.27±0.06

(n=6)

5-HIAA/5-HT 0.25±0.02

(n=6)

0.18±0.02

(n=5)

0.25±0.02

(n=6)

0.27±0.06

(n=6)

0.23±0.03

(n=8)

0.22±0.02

(n=7)

0.23±0.02

(n=6)

0.25±0.04

(n=6)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. In the same line, values with

same letter are significantly different. DA- dopamine; DOPAC- 3,4-dihydroxyphenylacetic acid; HVA-

homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid. a p<0.05.

Table 3.3.1.3.2- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the Nac, 30 minutes and 1 day after the end of cocaine exposure period.

30 min. 1 day

Male Female Male Female

saline cocaine saline cocaine saline cocaine saline cocaine

DOPAC/DA 0.28±0.06

(n=6)

0.24±0.03

(n=5)

0.25±0.04

(n=6)

0.19±0.02

(n=6)

0.24±0.02

(n=8)

0.29±0.04

(n=6)

0.33±0.04

(n=6)

0.27±0.05

(n=6)

HVA/DA 0.07±0.02

(n=6)

0.08±0.02

(n=6)

0.06±0.01

(n=6)

0.05±0.01

(n=6)

0.08±0.01

(n=8)

0.08±0.01

(n=6)

0.06±0.00

(n=6)

0.07±0.01

(n=6)

(DOPAC+HVA)/DA 0.36±0.08

(n=6)

0.32±0.05

(n=6)

0.30±0.05

(n=6)

0.24±0.02

(n=6)

0.32±0.02

(n=8)

0.37±0.04

(n=6)

0.39±0.04

(n=6)

0.34±0.06

(n=6)

5-HIAA/5-HT 0.50±0.21

(n=6)

0.40±0.19

(n=6)

0.54±0.20

(n=6)

0.40±0.18

(n=6)

0.32±0.12

(n=8)

0.73±0.26

(n=6)

0.79±0.32

(n=6)

0.52±0.21

(n=6)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid.

Within the dorsal striatum, no significant differences were detected in the levels of

DA and 5-HT and their metabolites between cocaine-treated animals and their respective

controls at both time points of analyses (figure 3.3.1.3.3). However, 1 day after the end of

the exposure period, the (DOPAC+HVA)/DA ratio in the dorsal striatum were higher in

cocaine-treated males than in control males [Z=-2.083 (2-tailed) p<0.05] (table 3.3.1.3.3).

Also, at this time point, the HVA/DA and (DOPAC+HVA)/DA ratios were higher in cocaine-

treated males than in cocaine-treated females, [Z=-2.286 (2-tailed) p<0.05] and [Z=-2.143

a a

98

(2-tailed) p<0.05], respectively. This sex effects was not observed in the control group. No

differences were found on 5-HT turnover between the experimental groups.

Dorsal striatum

A

0

1500

3000

4500

6000

7500

9000

DA DOPAC HVA DA DOPAC HVA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

30 min. 1 day

B

0

200

400

600

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

30 min. 1 day

Figure 3.3.1.3.3- Effects of chronic cocaine exposure throughout adolescence on the (A) levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the dorsal striatum,

determined by HPLC-EC for male and female rats, 30 minutes and 1 day after the last administration. Each

column represents the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for

at least 6 animals per group.

99

Table 3.3.1.3.3- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the dorsal striatum, 30 minutes and 1 day after the end of cocaine exposure period.

30 min. 1 day

Male Female Male Female

saline cocaine saline cocaine saline cocaine saline Cocaine

DOPAC/DA 0.18±0.01

(n=7)

0.24±0.02

(n=6)

0.21±0.03

(n=6)

0.20±0.02

(n=6)

0.19±0.01

(n=8)

0.30±0.06

(n=7)

0.17±0.02

(n=6)

0.18±0.01

(n=6)

HVA/DA 0.09±0.01

(n=7)

0.12±0.02

(n=6)

0.08±0.01

(n=6)

0.11±0.01

(n=6)

0.09±0.01

(n=8)

0.12±0.01

(n=7)

0.07±0.01

(n=6)

0.07±0.01

(n=6)

(DOPAC+HVA)/DA 0.27±0.02

(n=7)

0.35±0.04

(n=6)

0.29±0.04

(n=6)

0.30±0.02

(n=6)

0.28±0.01

(n=8)

0.42±0.07

(n=7)

0.24±0.03

(n=6)

0.26±0.02

(n=6)

5-HIAA/5-HT 2.14±1.05

(n=7)

1.73±1.10

(n=5)

1.62±1.15

(n=6)

2.17±1.11

(n=6)

1.16±0.76

(n=7)

1.21±0.83

(n=5)

1.57±1.11

(n=5)

2.52±1.13

(n=6)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. In the same line, values with

same letter are significantly different. DA- dopamine; DOPAC- 3,4-dihydroxyphenylacetic acid; HVA-

homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid. a,b,c p<0.05.

Within the prefrontal cortex, no statistical significant differences were found on the

levels of monoamines and their metabolites or their turnover rates (figure 3.3.1.3.4 and

table 3.3.1.3.4).

Within the hippocampus, no statistical significant differences were observed on the

levels of DA and its metabolite DOPAC, and on the levels of 5-HT and its metabolite 5-

HIAA between cocaine exposed and control males, as well as between cocaine exposed

and control females, both 30 minutes and 1 day after the last administration (figure

3.3.1.3.5). Also, no differences were found on DA and 5-HT turnover rates between the

experimental groups (table 3.3.1.3.5).

Hippocampal levels of HVA were not detectable by HPLC-EC in a considerable

number of samples. Therefore, in this brain region, only the levels of the DA metabolite

DOPAC was considered for the analysis.

a a,c

b b

c

100

Prefrontal cortex

A

0

20

40

60

80

100

DA DOPAC HVA DA DOPAC HVA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

30 min. 1 day

B

0

100

200

300

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

30 min. 1 day

Figure 3.3.1.3.4- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the prefrontal cortex,

determined by HPLC-EC for male and female rats, 30 minutes and 1 day after the last administration. Each

column represents the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for

at least 5 animals per group.

101

Hippocampus

A

0

5

10

15

20

DA DOPAC DA DOPAC

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

30 min. 1 day

B

0

200

400

600

800

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

Figure 3.3.1.3.5- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and on (B) the levels of 5-HT and its metabolite 5-HIAA in the hippocampus determined

by HPLC-EC for male and female rats, 30 minutes and 1 day after the last administration. Each column

represents the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for at least

6 animals per group.

102

Table 3.3.1.3.4- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the prefrontal cortex, 30 minutes and 1 day after the end of cocaine exposure period.

30 min. 1 day

Male Female Male Female

saline cocaine saline cocaine saline cocaine saline cocaine

DOPAC/DA 0.30±0.04

(n=7)

0.40±0.11

(n=5)

0.34±0.05

(n=6)

0.35±0.13

(n=5)

0.37±0.10

(n=8)

0.33±0.05

(n=7)

0.36±0.08

(n=6)

0.28±0.07

(n=6)

HVA/DA 0.61±0.09

(n=7)

0.54±0.14

(n=6)

0.51±0.08

(n=6)

0.58±0.12

(n=5)

0.80±0.14

(n=8)

0.58±0.09

(n=7)

0.45±0.09

(n=6)

0.47±0.07

(n=6)

(DOPAC+HVA)/DA 0.91±0.09

(n=7)

0.94±0.24

(n=6)

0.86±0.11

(n=6)

0.93±0.25

(n=5)

1.16±0.21

(n=8)

0.91±0.11

(n=7)

0.82±0.14

(n=6)

0.74±0.12

(n=6)

5-HIAA/5-HT 0.11±0.03

(n=7)

0.12±0.03

(n=6)

0.08±0.02

(n=6)

0.11±0.03

(n=5)

0.11±0.03

(n=8)

0.10±0.02

(n=7)

0.11±0.03

(n=6)

0.07±0.02

(n=6)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid.

Table 3.3.1.3.5- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA] and [5-HIAA]/[5-HT], in the

hippocampus, 30 minutes and 1 day after the end of cocaine exposure period.

30 min. 1 day

Male Female Male Female

saline cocaine saline cocaine saline cocaine saline cocaine

DOPAC/DA 0.54±0.14

(n=6)

0.83±0.27

(n=6)

1.09±0.44

(n=6)

0.67±0.12

(n=6)

0.97±0.50

(n=8)

0.75±0.23

(n=7)

0.59±0.11

(n=6)

1.10±0.29

(n=6)

5-HIAA/5-HT 0.25±0.07

(n=6)

0.21±0.05

(n=6)

0.38±0.14

(n=6)

0.26±0.08

(n=6)

0.30±0.05

(n=8)

0.31±0.04

(n=7)

0.26±0.03

(n=6)

0.25±0.07

(n=6)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid.

Within the amygdala, 1 day after withdrawal, the levels of DA were higher in

cocaine exposed females as compared to control females [Z=-2.082 (2-tailed) p<0.05]

(figure 3.3.1.3.6). At 30 minutes after the last administration, control males displayed

higher DOPAC/DA ratio as compared to control females [Z=-2.082 (2-tailed) p<0.05]

(table 3.3.1.3.6). Furthermore, at this time point of analysis, control males also displayed

higher levels of DOPAC as compared to control females that almost reached significance

[Z=-1.922 (2-tailed) p=0.055]. The statistical analysis also revealed, in control males, a

significant decrease on the levels of HVA from 30 minutes to 1 day after the last

administration [Z=-2.000 (2-tailed) p<0.05], and a decrease on the levels of DOPAC that

almost reached the statistical significance [Z=-1.936 (2-tailed) p=0.053], that were not

103

observed in treated animals. No differences were found on the levels of 5-HT and its

metabolite or its turnover between the experimental groups.

Amygdala

A

0

40

80

120

160

200

DA DOPAC HVA DA DOPAC HVA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

30 min. 1 day

B

0

100

200

300

400

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

30 min. 1 day

Figure 3.3.1.3.6- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the amygdala,

determined by HPLC-EC for male and female rats, 30 minutes and 1 day after the last administration. Each

column represents the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for

at least 6 animals per group. Columns with the same letter are significantly different from each other. a,b

p<0.05.

b

b

a a

104

Table 3.3.1.3.6- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the amygdala, 30 minutes and 1 day after the end of cocaine exposure period.

30 min. 1 day

Male Female Male Female

saline cocaine saline cocaine saline cocaine saline cocaine

DOPAC/DA 0.37±0.06

(n=6)

0.26±0.06

(n=5)

0.19±0.05

(n=6)

0.33±0.10

(n=6)

0.33±0.06

(n=8)

0.31±0.08

(n=7)

0.38±0.09

(n=6)

0.23±0.06

(n=6)

HVA/DA 0.18±0.06

(n=6)

0.26±0.13

(n=6)

0.14±0.04

(n=6)

0.08±0.03

(n=6)

0.06±0.10

(n=7)

0.11±0.05

(n=7)

0.08±0.03

(n=6)

0.06±0.02

(n=6)

(DOPAC+HVA)/DA 0.55±0.11

(n=6)

0.47±0.13

(n=6)

0.33±0.08

(n=6)

0.41±0.10

(n=6)

0.39±0.07

(n=7)

0.42±0.11

(n=7)

0.45±0.12

(n=6)

0.30±0.06

(n=6)

5-HIAA/5-HT 0.64±0.22

(n=6)

0.48±0.15

(n=6)

0.38±0.14

(n=6)

0.68±0.30

(n=6)

0.39±0.09

(n=8)

0.54±0.19

(n=7)

0.39±0.09

(n=6)

0.44±0.10

(n=6)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. In the same line, values with

same letter are significantly different. DA- dopamine; DOPAC- 3,4-dihydroxyphenylacetic acid; HVA-

homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid. a p<0.05.

Within the hypothalamus, the levels of 5-HIAA in cocaine exposed males

increased from 30 minutes to 1 day after the last administration [Z=-2.082 (2-tailed)

p=0.037] (figure 3.3.1.3.7). Moreover, 30 minutes after the last administration, cocaine

exposed males displayed lower 5-HIAA/5-HT ratio [Z=-2.714 (2-tailed) p<0.01] and an

increase in this ratio from 30 minutes to 1 day after the last administration [Z=-2.562 (2-

tailed) p=0.01] (table 3.3.1.3.7). No differences were found in this brain region on DA, its

metabolites or its turnover, between the experimental groups.

a a

105

Hypothalamus

A

DA DOPAC HVA DA DOPAC HVA

01020

100

200

300

400

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

B

0

100

200

300

400

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

30 min. 1 day

Figure 3.3.1.3.7- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the hypothalamus,

determined by HPLC-EC for male and female rats, 30 minutes and 1 day after the last administration. Each

column represents the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for

at least 5 animals per group. Columns with the same letter are significantly different from each other. a p<0.05

a a

30 min. 1 day

saline male cocaine male saline female cocaine female

106

Table 3.3.1.3.7- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the hypothalamus, 30 minutes and 1 day after the end of cocaine exposure period.

30 min. 1 day

Male Female Male Female

saline cocaine saline cocaine saline cocaine saline cocaine

DOPAC/DA 0.24±0.07

(n=7)

0.26±0.05

(n=6)

0.32±0.06

(n=6)

0.26±0.06

(n=6)

0.28±0.06

(n=7)

0.28±0.04

(n=6)

0.23±0.05

(n=5)

0.33±0.09

(n=5)

HVA/DA 0.03±0.01

(n=7)

0.03±0.01

(n=6)

0.03±0.01

(n=6)

0.04±0.01

(n=6)

0.03±0.01

(n=7)

0.06±0.03

(n=6)

0.01±0.00

(n=5)

0.03±0.02

(n=5)

(DOPAC+HVA)/DA 0.27±0.07

(n=7)

0.30±0.05

(n=6)

0.35±0.06

(n=6)

0.30±0.06

(n=6)

0.30±0.07

(n=7)

0.34±0.06

(n=6)

0.24±0.05

(n=5)

0.36±0.09

(n=5)

5-HIAA/5-HT 0.27±0.06

(n=7)

0.14±0.01

(n=6)

0.34±0.07

(n=6)

0.25±0.10

(n=6)

0.23±0.04

(n=7)

0.36±0.06

(n=6)

0.23±0.04

(n=5)

0.41±0.24

(n=5)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. In the same line, values with

same letter are significantly different. DA- dopamine; DOPAC- 3,4-dihydroxyphenylacetic acid; HVA-

homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid. a p<0.01, b p<0.02.

Within the dorsal raphe nucleus, statistical analysis revealed that there were no

significant differences on the levels of DA and its metabolites, DOPAC and HVA between

cocaine exposed and control male and female rats, both 30 minutes and 1 day after the

last administration (figure 3.3.1.3.8). At 30 minutes after the last administration, cocaine

exposed females displayed higher levels of 5-HT as compared to control females that

almost reached significance [Z=-1.922 (2-tailed) p=0.055]. And the levels of 5-HT

decreased in cocaine exposed females from 30 minutes to 1 day after the last

administration of cocaine [Z=-2.242 (2-tailed) p<0.05]. Also, in cocaine exposed females it

was observed an increase in the 5-HIAA/5-HT ratio, from 30 minutes to 1 day after the last

administration of cocaine [Z=-2.402 (2-tailed) p<0.02] (table 3.3.1.3.8). Statistical analysis

also revealed that 30 minutes after the last administration, the 5-HIAA/5-HT ratio was

lower in cocaine-treated males as compared to control males [Z=-2.402 (2-tailed) p<0.02]

(table 3.1.3.8). And, although not statistically significant, cocaine exposed male rats

presented an increase on the levels of 5-HIAA from 30 minutes to 1 day after the last

administration of cocaine [Z=-1.922 (2-tailed) p=0.055]. No differences were found in the

DA turnover in this brain area.

a a,b b

107

Dorsal raphe nucleus

A

0

50

100

150

200

DA DOPAC HVA DA DOPAC HVA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

30 min. 1 day

B

0

200

400

600

800

1000

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

30 min. 1 day

Figure 3.3.1.3.8- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the dorsal raphe

nucleus, determined by HPLC-EC for male and female rats, 30 minutes and 1 day after the last administration.

Each column represents the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of

protein, for at least 6 animals per group. Columns with the same letter are significantly different from each

other. a p<0.05.

a

a

108

Table 3.3.1.3.8- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the dorsal raphe nucleus, 30 minutes and 1 day after the end of cocaine exposure period.

30 min. 1 day

Male Female Male Female

saline cocaine saline cocaine saline cocaine saline cocaine

DOPAC/DA 0.85±0.23

(n=6)

0.70±0.22

(n=6)

1.03±0.13

(n=6)

1.12±0.27

(n=6)

1.16±0.26

(n=6)

0.75±0.17

(n=6)

1.10±0.24

(n=6)

1.00±0.26

(n=6)

HVA/DA 0.16±0.06

(n=6)

0.08±0.02

(n=6)

0.12±0.03

(n=6)

0.09±0.02

(n=6)

0.10±0.03

(n=6)

0.10±0.02

(n=6)

0.06±0.02

(n=6)

0.15±0.05

(n=6)

(DOPAC+HVA)/DA 1.01±0.26

(n=6)

0.78±0.22

(n=6)

1.15±0.11

(n=6)

1.21±0.27

(n=6)

1.23±0.28

(n=6)

0.85±0.18

(n=6)

1.17±0.25

(n=6)

1.15±0.28

(n=6)

5-HIAA/5-HT 0.28±0.02

(n=6)

0.20±0.02

(n=6)

0.26±0.02

(n=6)

0.20±0.02

(n=6)

0.34±0.05

(n=6)

0.27±0.02

(n=6)

0.26±0.02

(n=6)

3.40±3.11

(n=6)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. In the same line, values with

same letter are significantly different. DA- dopamine; DOPAC- 3,4-dihydroxyphenylacetic acid; HVA-

homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid. a,b p<0.02.

Within the cerebellum, statistical analysis revealed that there were no significant

difference on the levels of DA and its metabolites, DOPAC and HVA, and on the levels of

5-HT and 5-HIAA between cocaine exposed and control male and female rats, both at 30

minutes or 1 day after the last administration (figure 3.3.1.3.9). However, 1 day after the

last administration, the levels of 5-HT were lower in cocaine-treated males than in

cocaine-treated females [Z=-2.000 (2-tailed) p<0.05], and, although not statistically

significant, there was a trend to this same sex effect in saline-treated animals [Z=-1.936

(2-tailed) p=0.053]. In cocaine exposed males the DA and HVA levels decreased from 30

minutes to 1 day after the last administration (DA: [Z=-2.030 (2-tailed) p<0.05]; HVA: [Z=-

2.030 (2-tailed) p<0.05]). No differences were found both on DA and 5-HT turnover rates

between the experimental groups (table 3.3.1.3.9).

a a b b

109

Cerebellum

A

0

5

10

15

DA DOPAC HVA DA DOPAC HVA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

30 min. 1 day

B

0

20

40

60

80

100

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

30 min. 1 day

Figure 3.3.1.3.9- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the cerebellum,

determined by HPLC-EC for male and female rats, 30 minutes and 1 day after the last administration. Each

column represents the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for

at least 5 animals per group. Columns with the same letter are significantly different from each other. a,b,c

p<0.05.

c

c

a

a b b

110

Table 3.3.1.3.9- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the cerebellum, 30 minutes and 1 day after the end of cocaine exposure period.

30 min. 1 day

Male Female Male Female

saline cocaine saline cocaine saline cocaine saline cocaine

DOPAC/DA 0.83±0.24

(n=7)

0.62±0.20

(n=5)

0.75±0.16

(n=6)

0.72±0.13

(n=6)

0.70±0.18

(n=8)

0.90±0.23

(n=7)

0.77±0.14

(n=6)

0.79±0.22

(n=6)

HVA/DA 1.05±0.21

(n=7)

0.74±0.17

(n=5)

0.62±0.19

(n=6)

0.82±0.19

(n=6)

0.68±0.13

(n=8)

0.95±0.22

(n=7)

0.90±0.22

(n=6)

0.93±0.26

(n=6)

(DOPAC+HVA)/DA 1.88±0.31

(n=7)

1.36±0.23

(n=5)

1.38±0.22

(n=6)

1.54±0.26

(n=6)

1.38±0.14

(n=8)

1.85±0.33

(n=7)

1.67±0.23

(n=6)

1.72±0.34

(n=6)

5-HIAA/5-HT 0.23±0.03

(n=7)

0.18±0.05

(n=5)

0.18±0.02

(n=6)

0.18±0.01

(n=6)

0.22±0.03

(n=8)

0.48±0.25

(n=7)

0.15±0.01

(n=6)

0.15±0.02

(n=6)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid.

3.3.2. Behavioural studies and associated measures

3.3.2.1. Anxiety-like behaviour

Elevated plus maze test. On the PND 52, 2 days after the last administration of

cocaine, animals were tested in the EPM apparatus, in a five minute session. EPM

behavioural data was analysed by a 2-way ANOVA (sex x treatment). Differences

between specific groups were determined by post-hoc analyses using the Student’s t-test.

The behavioural category, risk-like behaviour, was analysed by the non-parametric Mann-

Whitney U Test.

Statistical analyses revealed a main effect of sex [F(1,29)=5.488; p<0.05], and an

interaction between sex and treatment [F(1,29)=4.810; p<0.05] in the number of entries

into the open arms. Males presented lower frequency of open arms entries than females

and post-hoc comparisons indicate that, within the cocaine group, males had lower

frequency than females [t(15)=-3.447; p<0.005] (figure 3.3.2.1.1 A). ANOVA results also

revealed an interaction between sex and treatment for the number of closed arms entries

[F(1,29)=5.517; p<0.03], with cocaine-treated males showing a trend towards decreased

entries into the closed arms as compared with cocaine-treated females [t(15)=-2.112;

p=0.052] (figure 3.3.2.1.1 C). For the total arm entries, statistical analysis revealed an

interaction between sex and treatment [F(1,29)=6.955; p<0.02], with a post-hoc

comparison showing that there was a decrease in the number of entries of cocaine-

treated males when compared with cocaine-treated females [t(15)=-3.468; p<0.005]

(figure 3.3.2.1.1 D).

111

No significant differences were found in the time spent in open and closed arms

between both cocaine- and saline-treated male and female rats (table 3.3.2.2.1).

ANOVA revealed an interaction between sex and treatment in the frequency of

entries into central platform [F(1,29)=7.340; p<0.02]. Within the cocaine-exposed group

there was a decrease in entries in males [t(15)=-3.570; p<0.005] (figure 3.3.2.1.3 A).

Within the male group, cocaine treatment decrease, although no significantly, the

frequency of central platform entries, [t(15)=-2.052; p=0.058] (figure 3.3.2.1.3 A).

Statistical analysis also revealed a interaction between sex and treatment in the time

spent in the central platform [F(1,29)= 9.606; p<0.005], a post hoc analysis using

student’s t-test shown that within the male group, cocaine significantly decreased the time

spent in the central platform [t(15)=-3.433; p<0.005], and that in cocaine exposed group

males spent less time in central platform than females [t(15)=-3.037; p<0.01] (figure

3.3.2.1.3 B).

A B

02468

10

male female

Num

ber o

f ent

ries

saline cocaine

0

20

40

60

male female

% o

f ent

ries

saline cocaine

C D

02468

10

male female

Num

ber o

f ent

ries

saline cocaine

05

101520

male female

Num

ber o

f ent

ries

saline cocaine

Figure 3.3.2.1.1- Effects of chronic cocaine exposure throughout adolescence in the number of arm entries

during the elevated plus maze test, performed 2 days after the last administration of cocaine, both by male

and female rats. A- Number of open arm entries; B- % of open arm entries; C- Number of closed arm entries;

D- Number of total arm entries. Each column represents the mean + S.E.M.. Saline males, n=8; Cocaine

males, n=9; saline females, n=8; cocaine females, n=8. a,b p<0.005.

Open arms Open arms

Closed arms Total arm entries

a

a

b

b

112

A B

0

50

100

150

male female

Dur

atio

n (s

ec.)

saline cocaine

0

80

160

240

male female

Dur

atio

n (s

ec.)

saline cocaine

Figure 3.3.2.1.2- Effects of chronic cocaine exposure throughout adolescence on the time spent (A) in the

open arms and (B) in the closed arms during the elevated plus maze test performed 2 days after the last

administration of cocaine both by male and female rats. Each column represents the mean + S.E.M.. Saline

males, n=8; cocaine males, n=9; saline females, n=8; cocaine females, n=8.

A B

05

101520

male female

Num

ber o

f ent

ries

saline cocaine

0

20

40

60

male female

Dur

atio

n (s

ec.)

saline cocaine

Figure 3.3.2.1.3- Effects of chronic cocaine exposure throughout adolescence (A) on the number of central

platform entries and (B) on the time spent in the central platform during the elevated plus maze test performed

2 days after the last administration of cocaine, both by male and female rats. Each column represents the

mean + S.E.M. Saline males, n=8; cocaine males, n=9; saline females, n=8; cocaine females, n=8. Columns

with the same letter are significantly different from each other. a,b, p<0.005, c p<0.01.

Importantly, non-parametric statistical analysis revealed that within the male group

cocaine enhanced the risk-like behaviour (jump of the apparatus and walk on top of

closed arms) [Z=-2.785 (2-tailed) p<0.005] (table 3.3.2.1.1). This behavioural category

was only observed in cocaine-treated males. ANOVA also revealed a treatment effect in

the frequency [F(1,29)=8.295; p<0.01] and in the time spent [F(1,29)=5.455; p<0.05]

performing head-dipping behaviour. Cocaine administration increased the frequency and

the time spent in the head dipping behaviour (table 3.3.2.1.1). No group differences were

detected in frequency and duration of rearing and grooming during the EPM.

Central platform

Open arms Closed arms

Central platform

a

a b

b,c

c

113

Table 3.3.2.1.1- Frequency and duration of behavioural categories like risk-like behaviour, rearing, head

dipping and grooming, during the 5 minutes of the elevated plus maze test, in saline- and cocaine-treated

male and female rats.

Male Female

saline

(n=8) cocaine

(n=9) saline (n=8)

cocaine (n=8)

Risk-like Behaviour frequency 0.00±0.00 0.67±0.17 0.00±0.00 0.00±0.00

duration (sec.) - - - -

Rearing frequency 16.38±1.21 15.56±2.61 17.38±1.92 21.00±2.67

duration (sec.) 39.50±5.33 39.04±7.98 47.02±5.47 56.30±8.23

Head-dipping frequency 2.50±0.76 5.22±1.40 3.75±0.92 8.50±2.25

duration (sec.) 1.68±0.51 9.48±6.61 2.77±0.89 8.07±2.74

Grooming frequency 2.00±0.68 2.56±0.82 2.63±0.65 1.75±0.56

duration (sec.) 14.50±6.92 13.57±4.93 14.88±5.68 6.27±2.74

Data expressed as the mean value ± S.E.M.; n, number of animals per group. In the same line, values with

same letter are significantly different. a,b p<0.02.

Corticosterone plasma levels. After performing the EPM animals were decapitated

and corticosterone levels were measured in the obtained plasma. Statistical significance

between groups was assessed using the non-parametric Mann-Whitney U Test.

No significant differences were observed on plasma levels of corticosterone

between cocaine-treated and saline control males, and between cocaine-treated and

saline control females, after the EPM. However, nonparametric Mann-Whitney U test

showed that cocaine-treated males had lower levels of plasma corticosterone than

cocaine-treated females [Z=-2.556 (2-tailed) p<0.02] (figure 3.3.2.1.4).

0100200300400

male female

Cor

ticos

tero

ne

leve

ls (n

g/m

L)

saline cocaine

Figure 3.3.2.1.4- Effects of chronic cocaine exposure throughout adolescence on the alterations of the

corticosterone plasma levels of corticosterone induced by the elevated plus maze test, both in male and

female rats. Each column represents the mean + S.E.M for 6 animals per group. Columns with the same letter

are significantly different from each other. a p<0.02.

a

a

a a,b b

114

Neurochemical determinations. The levels of DA, 5-HT and their metabolites,

DOPAC, HVA and 5-HIAA were measured by HPLC-EC, in the prefrontal cortex,

hippocampus, amygdala, hypothalamus, SN-VTA and in the dorsal raphe nucleus. The

obtained data were analyzed by the non-parametric Mann-Whitney U Test.

Within the SN-VTA, no significant differences were detected on the levels of DA

and 5-HT, their metabolites and their turnover between the experimental groups (figure

3.3.2.1.9 and table 3.3.2.1.6).

SN-VTA

A

0

200

400

600

800

DA DOPAC HVANeu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

B

0

150

300

450

600

750

5-HT 5-HIAANeu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

Figure 3.3.2.1.9- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the SN-VTA after the

elevated plus maze test, determined by HPLC-EC for male and female rats. Each column represents the

mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for at least 5 animals per

group.

115

Table 3.3.2.1.6- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the SN-VTA, in cocaine- and saline-treated male and female rats, after the elevated plus

maze test.

Male Female

saline cocaine saline cocaine

DOPAC/DA 0.32±0.10

(n=5)

0.31±0.05

(n=6)

0.35±0.08

(n=6)

0.34±0.07

(n=6)

HVA/DA 0.10±0.03

(n=5)

0.11±0.03

(n=6)

0.11±0.02

(n=6)

0.11±0.01

(n=6)

(DOPAC+HVA)/DA 0.43±0.12

(n=5)

0.42±0.08

(n=6)

0.45±0.09

(n=6)

0.45±0.08

(n=6)

5-HIAA/5-HT 0.43±0.28

(n=5)

0.27±0.11

(n=6)

0.19±0.03

(n=6)

0.21±0.02

(n=6)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid.

Within the dorsal raphe nucleus no differences were observed on the levels of DA

and 5-HT and their metabolites, and on DA and 5-HT turnover rates between the

experimental groups (figure 3.3.2.1.10 and table 3.3.2.1.7).

Within the prefrontal cortex there were no significant differences on the levels of

DA and 5-HT and their metabolites between the experimental groups (figure 3.3.2.1.6).

However, the HVA/DA turnover rate was increased in cocaine-treated males as compared

to control males [Z=-2.191 (2-tailed) p<0.05] and to cocaine-treated females [Z=-2.191 (2-

tailed) p<0.05]. The 5-HT turnover rate was also increased in cocaine-treated males as

compared to control males [Z=-2.008 (2-tailed) p=0.045] (table 3.3.2.1.3).

116

Dorsal raphe nucleus

A

0

30

60

90

120

DA DOPAC HVANeu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

B

0

250

500

750

1000

5-HT 5-HIAANeu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

Figure 3.3.2.1.10- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and

its metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the dorsal raphe

nucleus after the elevated plus maze test, determined by HPLC-EC for male and female rats. Each column

represents the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for at least

5 animals per group.

117

Prefrontal cortex

A

0

15

30

45

60

DA DOPAC HVANeu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

B

0

100

200

300

400

500

5-HT 5-HIAANeu

rotr

ansm

ittte

rs c

once

ntra

tion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

Figure 3.3.2.1.6- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the prefrontal cortex

after the elevated plus maze test, determined by HPLC-EC for male and female rats. Each column represents

the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for at least 5 animals

per group.

118

Table 3.3.2.1.7- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the dorsal raphe nucleus, in cocaine- and saline-treated male and female rats, after the

elevated plus maze test.

Male Female

saline cocaine saline cocaine

DOPAC/DA 0.64±0.07

(n=5)

0.63±0.07

(n=6)

0.83±0.18

(n=6)

0.88±0.13

(n=6)

HVA/DA 0.09±0.03

(n=5)

0.10±0.03

(n=6)

0.14±0.03

(n=6)

0.12±0.03

(n=6)

(DOPAC+HVA)/DA 0.73±0.08

(n=5)

0.74±0.09

(n=6)

0.97±0.20

(n=6)

1.00±0.14

(n=6)

5-HIAA/5-HT 0.20±0.03

(n=5)

0.18±0.02

(n=6)

0.26±0.04

(n=6)

0.27±0.06

(n=6)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid.

Table 3.3.2.1.3- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the prefrontal cortex, in cocaine- and saline-treated male and female rats, after the

elevated plus maze test.

Male Female

saline cocaine saline cocaine

DOPAC/DA 1.57±0.11

(n=6)

1.86±0.48

(n=5)

1.57±0.31

(n=6)

1.29±0.13

(n=6)

HVA/DA 0.61±0.07

(n=6)

0.80±0.03

(n=5)

0.51±0.10

(n=6)

0.68±0.04

(n=6)

(DOPAC+HVA)/DA 2.17±0.15

(n=6)

2.67±0.50

(n=5)

2.08±0.27

(n=6)

1.97±0.16

(n=6)

5-HIAA/5-HT 0.06±0.00

(n=6)

0.09±0.01

(n=5)

0.11±0.04

(n=6)

0.07±0.02

(n=6)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. In the same line, values with

same letter are significantly different. DA- dopamine; DOPAC- 3,4-Dihydroxyphenylacetic acid; HVA-

Homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid. a,b,c p<0.05.

In the hippocampus there were no differences on the levels of DA and 5-HT and

their metabolites between the experimental groups (figure 3.3.2.1.7). However, the

DOPAC/DA turnover rate was decreased in cocaine-treated females as compared to

control females [Z=-2.008 (2-tailed) p<0.05] (table 3.2.1.4). Cocaine-treated females also

displayed lower DOPAC/DA turnover rate as compared to cocaine-treated males that

almost reached significance [Z=-1.925 (2-tailed) p=0.054]. No differences were found on

5-HT turnover rate between the experimental groups (table 3.3.2.1.4).

a a,b

c c

b

119

Hippocampus

A

0

10

20

30

40

DA DOPACNeu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

B

0

100

200

300

400

5-HT 5-HIAANeu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

Figure 3.3.2.1.7- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolite DOPAC and on (B) the levels of 5-HT and its metabolite 5-HIAA in the hippocampus after the

elevated plus maze test, determined by HPLC-EC for male and female rats. Each column represents the

mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for at least 5 animals per

group.

120

Table 3.3.2.1.4- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA] and [5-HIAA]/[5-HT], in the

hippocampus, in cocaine- and saline-treated male and female rats, after the elevated plus maze test.

Male Female

saline cocaine saline cocaine

DOPAC/DA 2.57±0.37

(n=6)

2.31±0.22

(n=6)

2.73±0.53

(n=5)

1.76±0.12

(n=6)

5-HIAA/5-HT 0.19±0.03

(n=6)

0.20±0.03

(n=6)

0.21±0.04

(n=5)

0.27±0.05

(n=6)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. In the same line, values with

same letter are significantly different. DA- dopamine; DOPAC- 3,4-dihydroxyphenylacetic acid; 5-HT-

serotonin; 5-HIAA- 5-hydroxyindolacetic acid. a p<0.05.

Within the amygdala, statistical analysis revealed that the levels of HVA in

cocaine-treated males were higher than in control males [Z=-2.242 (2-tailed) p<0.05]. No

significant difference were found on the levels of DA and its metabolite DOPAC, and on

the levels of 5-HT and 5-HIAA between cocaine exposed and control males, and between

cocaine exposed and control females after the elevated plus maze test (figure 3.3.2.1.5).

No differences were found both on DA and 5-HT turnover rates between the experimental

groups (table 3.3.2.1.2).

Within the hypothalamus, although there were no significant differences on the

levels of DA and 5-HT and their metabolites between the experimental groups (figure

3.3.2.1.8), cocaine-treated males displayed higher levels of DA than control males that

almost reached significance [Z=-1.924 (2-tailed) p=0.055]. No significant differences were

found on DA and 5-HT turnover rates between the experimental groups (table 3.3.2.1.5).

a a

121

Amygdala

A

0

50

100

150

200

DA DOPAC HVANeu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

B

0

150

300

450

600

5-HT 5-HIAANeu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

Figure 3.3.2.1.5- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the amygdala after

the elevated plus maze test, determined by HPLC-EC for male and female rats. Each column represents the

mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for 6 animals per group.

Columns with the same letter are significantly different from each other. a p<0.05

a a

122

Hypothalamus

A

0

100

200

300

400

DA DOPAC HVANeu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

B

0

100

200

300

400

500

5-HT 5-HIAANeu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

Figure 3.3.2.1.8- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the hypothalamus

after the elevated plus maze test, determined by HPLC-EC for male and female rats. Each column represents

the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for 6 animals per

group.

123

Table 3.3.2.1.2- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the amygdala, in saline- and cocaine-treated male and female rats, after the elevated plus

maze test.

Male Female

saline cocaine saline cocaine

DOPAC/DA 0.86±0.27

(n=6)

0.57±0.14

(n=6)

0.73±0.09

(n=6)

0.56±0.06

(n=6)

HVA/DA 0.17±0.10

(n=6)

0.14±0.02

(n=6)

0.10±0.02

(n=6)

0.15±0.02

(n=6)

(DOPAC+HVA)/DA 1.03±0.37

(n=6)

0.71±0.16

(n=6)

0.84±0.08

(n=6)

0.71±0.08

(n=6)

5-HIAA/5-HT 0.45±0.20

(n=6)

0.44±0.30

(n=6)

0.19±0.04

(n=6)

0.16±0.02

(n=6)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid.

Table 3.3.2.1.5- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the hypothalamus, in cocaine- and saline-treated male and female rats, after the elevated

plus maze test.

Male Female

saline cocaine saline cocaine

DOPAC/DA 0.38±0.07

(n=6)

0.43±0.09

(n=6)

0.54±0.09

(n=6)

0.50±0.09

(n=6)

HVA/DA 0.06±0.03

(n=6)

0.04±0.01

(n=6)

0.05±0.01

(n=6)

0.06±0.01

(n=6)

(DOPAC+HVA)/DA 0.44±0.08

(n=6)

0.47±0.10

(n=6)

0.60±0.09

(n=6)

0.55±0.10

(n=6)

5-HIAA/5-HT 0.17±0.04

(n=6)

0.20±0.03

(n=6)

0.19±0.06

(n=6)

0.24±0.06

(n=6)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid.

124

3.3.2.2. Depressive-like behaviour

Forced swim test. Starting 2 days after the last administration of cocaine, both

male and female Wistar rats performed a FST. Behavioural data was analysed by a 2-way

ANOVA with treatment (cocaine or saline) and sex as between-subject fixed-factors, and

behavioural measurements as the dependent variables.

Statistical analyses revealed an main effect of sex in the latency to struggling

behaviour [F(1,31)=4.522; p<0.05]. The latency to struggling was lower in males than in

females. No other significant behavioural differences were found in the FST between

experimental groups (table 3.3.2.2.1).

Table 3.3.2.2.1- Behaviour of male and female rats in a forced swim test after chronic cocaine exposure

during adolescence.

Male Female

saline n=9

cocaine n=9

saline n=9

cocaine n=8

Struggling

frequency 1.00±0.00 1.00±0.00 1.00±0.00 1.00±0.00

duration (sec.) 47.78±14.51 27.85±8.40 37.60±7.53 38.23±8.73

latency (sec.) 0.88±0.33 1.28±0.53 2.09±0.67 5.18±2.43

Immobility

frequency 0.00±0.00 0.00±0.00 0.00±0.00 0.00±0.00

duration (sec.) - - - -

latency (sec.) - - - -

Diving

frequency 0.44±0.34 1.22±1.00 0.44±0.34 0.50±0.50

duration (sec.) 0.37±0.36 1.53±1.29 0.81±0.54 0.68±0.68

latency (sec.) 253.40±30.91 253.04±31.33 252.66±31.87 269.74±30.27

Right rotation

frequency 28.67±2.48 31.22±2.52 34.44±4.60 35.34±3.08

duration (sec.) - - - -

latency (sec.) 4.85±2.48 3.36±0.71 4.12±1.67 1.57±0.62

Left rotation

frequency 36.56±2.91 32.44±3.22 33.78±4.02 28.88±3.95

duration (sec.) - - - -

latency (sec.) 2.13±0.93 3.36±1.49 4.81±1.56 3.75±1.83

Data expressed as the mean value ± S.E.M.; n, number of animals per group. In the same line, values with

same letter are significantly different. sec.- seconds.

125

Corticosterone plasma levels. At the end of FST animals were decapitated and

corticosterone levels were measured in the obtained plasma. Statistical significance

between groups was assessed using the non-parametric Mann-Whitney U Test. Statistical

analysis revealed no significant differences in the levels of corticosterone after the FST

between the experimental groups (figure 3.3.2.2.1).

0150300450600

male female

Cor

ticos

tero

ne

leve

ls (n

g/m

L)

saline cocaine

Figure 3.3.2.2.1- Effects of chronic cocaine exposure throughout adolescence on the alterations on the

plasma levels of corticosterone induced by a forced swim test, both in male and female rats. Each column

represents the mean + S.E.M for 6 animals per group.

Neurochemical determinations. The levels of DA, 5-HT and their metabolites,

DOPAC, HVA and 5-HIAA were measured by HPLC-EC, in the SN-VTA, dorsal raphe

nucleus, prefrontal cortex, hippocampus, amygdala and hypothalamus. The data obtained

was analyzed by the non-parametric Mann-Whitney U Test.

Within the SN-VTA, no significant differences were observed on the levels of DA

and 5-HT and their metabolites between the experimental groups (figure 3.3.2.2.6).

However, cocaine-treated males exhibited higher DOPAC/DA and (DOPAC+HVA)/DA

turnover rates as compared to control males (DOPAC/DA: [Z=-2.193 (2-tailed) p<0.05];

(DOPAC+HVA)/DA: [Z=-1.984 (2-tailed) p<0.05]) (table 3.3.2.2.6). No differences were

observed in the 5-HT turnover rate between the experimental groups.

Within the dorsal raphe nucleus, no differences were detected on the levels of DA

and 5-HT and their metabolites, and on DA and 5-HT turnover between the experimental

groups (figure 3.3.2.2.7 and table 3.3.2.2.7).

126

SN-VTA

A

0

100

200

300

400

500

DA DOPAC HVANeu

rotr

ansm

itter

s co

ncen

trat

ion

)ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

B

0

200

400

600

800

5-HT 5-HIAANeu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

Figure 3.3.2.2.6- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the SN-VTA after a

forced swim test, determined by HPLC-EC for male and female rats. Each column represents the mean +

S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for at least 5 animals per group.

127

Dorsal raphe nucleus

A

0

30

60

90

120

DA DOPAC HVANeu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

B

0

200

400

600

800

5-HT 5-HIAANeu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

Figure 3.3.2.2.7- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the dorsal raphe

nucleus after a forced swim test, determined by HPLC-EC for male and female rats. Each column represents

the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for at least 5 animals

per group.

128

Table 3.3.2.2.6- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the SN-VTA, in cocaine- and saline-treated male and female rats, after a forced swim test.

Male Female

saline cocaine saline cocaine

DOPAC/DA 0.30±0.05

(n=5)

0.46±0.05

(n=5)

0.27±0.05

(n=6)

0.39±0.07

(n=6)

HVA/DA 0.09±0.01

(n=5)

0.09±0.02

(n=5)

0.07±0.01

(n=6)

0.11±0.02

(n=6)

(DOPAC+HVA)/DA 0.39±0.05

(n=5)

0.55±0.04

(n=5)

0.34±0.06

(n=6)

0.50±0.09

(n=6)

5-HIAA/5-HT 0.14±0.01

(n=5)

0.17±0.05

(n=5)

0.20±0.03

(n=6)

0.20±0.05

(n=6)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. In the same line, values with

same letter are significantly different. DA- dopamine; DOPAC- 3,4-dihydroxyphenylacetic acid; HVA-

homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid. a,b p<0.05.

Table 3.3.2.2.7- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the dorsal raphe nucleus, in cocaine- and saline-treated male and female rats, after a

forced swim test.

Male Female

saline cocaine saline cocaine

DOPAC/DA 0.73±0.11

(n=6)

0.44±0.11

(n=6)

0.54±0.12

(n=5)

0.71±0.10

(n=6)

HVA/DA 0.13±0.04

(n=6)

0.16±0.03

(n=6)

0.15±0.04

(n=5)

0.15±0.03

(n=6)

(DOPAC+HVA)/DA 0.86±0.13

(n=6)

0.60±0.13

(n=6)

0.69±0.14

(n=5)

0.86±0.10

(n=6)

5-HIAA/5-HT 0.21±0.04

(n=6)

0.14±0.01

(n=6)

0.19±0.02

(n=5)

0.28±0.08

(n=6)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid.

Within the prefrontal cortex, also no differences were detected on the levels of DA

and 5-HT, their metabolites and their turnover rates between the experimental groups

(figure 3.3.2.2.3 and table 3.3.2.2.3).

a a

b b

129

Prefrontal cortex

A

0

10

20

30

40

DA DOPAC HVANeu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

B

0

50

100

150

200

250

5-HT 5-HIAANeu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

Figure 3.3.2.2.3- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the prefrontal cortex

after a forced swim test, determined by HPLC-EC for male and female rats. Each column represents the mean

+ S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for 6 animals per group.

130

Table 3.3.2.2.3- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the prefrontal cortex, in cocaine- and saline-treated male and female rats, after a forced

swim test.

Male Female

saline cocaine saline cocaine

DOPAC/DA 1.88±0.50

(n=6)

1.75±0.23

(n=6)

1.66±0.44

(n=6)

1.32±0.29

(n=6)

HVA/DA 0.93±0.10

(n=6)

0.89±0.17

(n=6)

0.78±0.08

(n=6)

0.69±0.10

(n=6)

(DOPAC+HVA)/DA 2.82±0.58

(n=6)

2.64±0.38

(n=6)

2.39±0.46

(n=6)

2.01±0.30

(n=6)

5-HIAA/5-HT 0.06±0.01

(n=6)

0.07±0.01

(n=6)

0.07±0.02

(n=6)

0.06±0.01

(n=6)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid.

Within the hippocampus, also no differences were detected on the levels of DA

and 5-HT, their metabolites and their turnover between the experimental groups (figure

3.3.2.2.4 and table 3.3.2.2.4).

Within the amygdala, no significant differences were detected on the levels of DA

and 5-HT and their metabolites (figure 3.3.2.2.2). However, control females displayed

increased levels of 5-HT [Z =-1.922 (2-tailed) p=0.055] and decreased (DOPAC+HVA)/DA

turnover rate [Z =-1.922 (2-tailed) p=0.055] when compared with control males that almost

reached significance [Z =-1.922 (2-tailed) p=0.055]. Furthermore, it was also observed a

lower 5-HIAA/5-HT rate in control females as compared to control males [Z =-2.242 (2-

tailed) p<0.05] (table 3.3.2.2.2). These sex effects were not observed in cocaine-treated

rats. Also in this brain region, although not significantly, cocaine-treated males exhibited

lower DOPAC/DA turnover rate as compared to control males [Z =-1.922 (2-tailed)

p=0.055].

131

Hippocampus

A

0

10

20

30

40

DA DOPACNeu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

B

0

50

100

150

200

250

5-HT 5-HIAANeu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

Figure 3.3.2.2.4- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolite DOPAC and on (B) the levels of 5-HT and its metabolite 5-HIAA in the hippocampus after a forced

swim test, determined by HPLC-EC for male and female rats. Each column represents the mean + S.E.M. of

monoamine levels, expressed as ng of monoamine/mg of protein, for at least 5 animals per group.

132

Amygdala

A

0

25

50

75

100

DA DOPAC HVANeu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

B

0

100

200

300

5-HT 5-HIAANeu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

Figure 3.3.2.2.2- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the amygdala after a

forced swim test, determined by HPLC-EC for male and female rats. Each column represents the mean +

S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for 6 animals per group.

133

Table 3.3.2.2.4- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA] and [5-HIAA]/[5-HT], in the

hippocampus, in cocaine- and saline-treated male and female rats, after a forced swim test.

Male Female

saline cocaine saline cocaine

DOPAC/DA 2.32±0.56

(n=6)

2.47±0.21

(n=6)

1.88±0.37

(n=5)

2.03±0.41

(n=6)

5-HIAA/5-HT 0.16±0.04

(n=6)

0.13±0.02

(n=6)

0.19±0.03

(n=5)

0.23±0.05

(n=6)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid.

Table 3.3.2.2.2- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the amygdala, in cocaine- and saline-treated male and female rats, after a forced swim

test.

Male Female

saline cocaine saline cocaine

DOPAC/DA 0.76±0.11

(n=6)

0.57±0.06

(n=6)

0.53±0.09

(n=6)

0.55±0.08

(n=6)

HVA/DA 0.13±0.04

(n=6)

0.09±0.03

(n=6)

0.05±0.02

(n=6)

0.07±0.03

(n=6)

(DOPAC+HVA)/DA 0.89±0.12

(n=6)

0.66±0.07

(n=6)

0.58±0.09

(n=6)

0.63±0.06

(n=6)

5-HIAA/5-HT 0.23±0.05

(n=6)

0.16±0.03

(n=6)

0.10±0.01

(n=6)

0.14±0.02

(n=6)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. In the same line, values with

same letter are significantly different. DA- dopamine; DOPAC- 3,4-dihydroxyphenylacetic acid; HVA-

homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid. a p<0.05.

Within the hypothalamus, after the FST no differences were detected on the levels

of DA and 5-HT, their metabolites, and on DA and 5-HT turnover between the

experimental groups (figure 3.3.2.2.5 and table 3.3.2.2.5).

a a

134

Hypothalamus

A

0

100

200

300

400

DA DOPAC HVANeu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

B

0

150

300

450

600

5-HT 5-HIAANeu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

Figure 3.3.2.2.5- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the hypothalamus

after a forced swim test, determined by HPLC-EC for male and female rats. Each column represents the mean

+ S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for 6 animals per group.

135

Table 3.3.2.2.5- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the hypothalamus, in cocaine- and saline-treated male and female rats, after a forced

swim test.

Male Female

saline cocaine saline cocaine

DOPAC/DA 0.53±0.12

(n=6)

0.43±0.12

(n=6)

0.33±0.04

(n=6)

0.47±0.19

(n=6)

HVA/DA 0.07±0.02

(n=6)

0.04±0.01

(n=6)

0.04±0.01

(n=6)

0.06±0.01

(n=6)

(DOPAC+HVA)/DA 0.60±0.13

(n=6)

0.48±0.13

(n=6)

0.37±0.06

(n=6)

0.53±0.10

(n=6)

5-HIAA/5-HT 0.17±0.04

(n=6)

0.14±0.03

(n=6)

0.13±0.03

(n=6)

0.23±0.06

(n=6)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid.

136

3.4. Discussion

3.4.1. Effects of chronic cocaine treatment during adolescence on the body weight evolution

It is known that any weight reduction gain during adolescence is likely to alter the

pace of development, delaying puberty, extending the adolescence period, and perhaps

altering the normal trajectory of developmental processes (Spear, 2007). Our data

showed that male rats presented higher weight gain than did females. In fact, it is known

that sex differences in body weight appear during adolescence (Macri et al., 2002).

However, body weight evolution was not affected by the chronic cocaine administration

both in male and female adolescent rats. These findings contrast with the known anorectic

and stress-like effects of cocaine. Cocaine decreases food intake and increases metabolic

processes through the activation of the sympathetic nervous system (Spector et al., 1972;

Balopole et al., 1979). Moreover, cocaine suppresses food intake in a dose-dependent

manner, with a dose between 10 to 30 mg/kg showing to be anorectic (Cooper and van

der Hoek, 1993). On the other hand, Laviola et al (1995) have shown that chronic cocaine

(10 or 20 mg/kg/day for 4 days) reduced body weight and food consumption in adult male

rats but not in periadolescents (PND 34-39) of either sex. Giorgetti and Zhdanova (2000)

also showed that adolescent rats chronically treated with cocaine (15 mg/kg i.p., twice a

day for 9 days) did not revealed any significant alteration either in the total amount of food

consumed over a 24-h period or in the body weight. These, as well as our findings,

suggest an age effect regarding cocaine effects on body weight evolution. In addition, it

was demonstrated that adolescent rats continue to gain body weight better than adults

during both cold and restraint stress (Gomez and Dallman, 2001; Gomez et al., 2002). It

has been proposed that, facing different types of chronic stress such as cold and restrain,

adolescent rats put their resources into ponderal growth, whereas adult rats protect the

reproductive capability (Gomez and Dallman, 2001; Gomez et al., 2002). This strategy

could also underlie the absence of body weight differences between cocaine exposed and

control adolescent animals observed in this study.

3.4.2. Effects of chronic cocaine treatment during adolescence on the neuroendocrine function

The acute stimulant effects of cocaine in the HPA axis in rats and in rhesus

monkeys is well known (Rivier and Vale, 1987; Borowsky and Kuhn, 1991; Sarnyai et al.,

1992; Sarnyai et al., 1996). The stimulatory cocaine effect in human HPA axis has also

been demonstrated (Mendelson et al., 1992; Teoh et al., 1994). However, there are

137

conflicting reports in the literature as to whether chronic cocaine exhibits persistent

stimulatory effects on the HPA axis. Tolerance to the stimulatory effects of cocaine has

also been reported in rats following chronic binge pattern of administration (Zhou et al.,

1996). This tolerance has been observed as an attenuated ACTH and corticosterone

responses to cocaine (Zhou et al., 1996; Zhou et al., 2003). In the present study, male

rats displayed an activation of the HPA axis in the last day of chronic cocaine treatment.

Thirty minutes after the last administration of cocaine the ACTH levels were increased in

males. However, the increased levels of ACTH did not result in the expected subsequent

increase in the levels of corticosterone. This could be explained by an altered adrenal

sensitization to the ACTH stimulatory.

A sex difference regarding HPA axis activation following cocaine was found 30

minutes after the last administration. Contrarily to what was observed in males, in females

no differences were seen in the ACTH or corticosterone plasma levels. The sex

differences in the HPA axis function are well known. Former findings have shown greater

corticosterone and ACTH levels in female, compared with male rats, both at baseline

(Atkinson and Waddell, 1997) and following a stress response (Rivier, 1993; Handa et al.,

1994; Yoshimura et al., 2003). Moreover, it is known that males and females display a

different HPA axis response to cocaine. However, contrarily to our data, most reports

describe that female rats exhibit a greater HPA axis activation following cocaine

administration (Kuhn and Francis, 1997).

Available research indicates that sex differences in the HPA axis function emerge

over adolescence, a phenomenon reflecting the concomitant initiation of regulatory effects

of sex hormones (reviewed by McCormick and Mathews, 2007). The ACTH responses to

cocaine are absent before puberty, when levels of gonadal steroids are fairly low (Kuhn

and Francis, 1997). In addition, ovariectomy decreased the ACTH response to cocaine in

females, whereas castration did not influence the response in males (Kuhn and Francis,

1997). Moreover, estrogen treatment of ovariectomized females increases CRH

production in the hypothalamus, ACTH and corticosterone production when facing a

stressor (Lesniewska et al., 1990). Based on these data, the failure of cocaine to induce

activation of the HPA axis in adolescent females could be explained by an immature

regulation of the HPA axis activity by gonadal hormones.

In the present study data from female rats were used without regarded hormonal

fluctuations. This strategy was based on the assumption that variations in gonadal steroid

hormone levels in the physiological range through the estrous cycle would produce

smaller changes than those that exist between males and females (Kuhn and Francis,

1997). In fact, several studies using females without regard to estrous state demonstrate

138

robust gender differences in the HPA axis function (Lesniewska et al., 1990; Aloisi et al.,

1994; Chisari et al., 1995).

Interestingly, 1 day after the last administration the altered HPA axis activity was

no longer present. No differences were observed in the levels of ACTH or corticosterone

between cocaine and saline exposed male or female rats. In fact, the levels of ACTH in

cocaine exposed males did not differ between 30 minutes and 1 day after the last

administration, while the levels of ACTH in control males increased between this two

assessment time points. This may indicate that the stress caused by the binge pattern of

administration induced a decrease in plasma levels of ACTH in control males, which was

not observed in cocaine males, most likely due to the acute effect of the 3rd daily cocaine

injection on HPA axis activation. In fact, it is known that repeated stress induces

alterations in the HPA axis function. For instance, Gomez et al. (2002) reported that in

adolescent male rats, beside the habituation of ACTH responses to repeated restraint

stress across days, a clear habituation of ACTH responses to restraint occurs within three

repetitions of the stress.

Nevertheless, the absence of HPA activation, both in male and female rats, 1 day

following the last administration of cocaine was unexpected. It has been demonstrated

with a variety of paradigms that withdrawal from cocaine induces an activation of the HPA

axis. However, Mantsch et al. (2007), using adult rats, demonstrated that plasma

corticosterone and CRH mRNA levels in the paraventricular nuclei of hypothalamus were

not altered 24 hours after the final administration of chronic cocaine (30mg/kg/day, for 14

days). These findings are also consistent with reports showing that basal corticosterone

levels were unaltered after 1 day of withdrawal from chronic cocaine (Levy et al., 1994;

Zorrilla et al., 2001; Baumann et al., 2004). However, Zhou et al. (2003) reported that 1

day after chronic binge administration (14 days of 3x15mg/kg i.p.) the levels of ACTH and

corticosterone were increased in adult male rats, although the hypothalamic CRH mRNA

levels were not different from that of saline controls.

However, age-related discontinuities in the response of the HPA axis to both stress

and psychostimulants have been demonstrated (Adriani and Laviola, 2000; Laviola et al.,

2002). Upon amphetamine administration, periadolescents failed to show an increase

across the course of the session in corticosterone release, that was observed in adults

(Adriani and Laviola, 2000). Moreover, it has been demonstrated that basal corticosterone

levels are elevated in adolescents compared to adult rodents (Laviola et al., 1999). This

elevated baseline is accompanied by an attenuated percentage increase in corticosterone

levels in response to psychostimulants (Adriani and Laviola, 2000). Thus, although

139

adolescents operate at a higher baseline, their HPA axis response to psychostimulants

may be proportionately masked (Schramm-Sapyta et al., 2006).

The age-related discontinuity in the HPA axis function may be underlying the

absence of HPA axis activation on day 1 from withdrawal in adolescent rats, observed in

our study. Moreover, this observation points to a reduced HPA axis response to chronic

binge cocaine administration during adolescence, suggesting that this axis is somewhat

hyporesponsive to cocaine during adolescence.

Adrenal weight, normalized to body weight, was not affected by the cocaine

exposure during adolescence since no differences were observed between cocaine-

treated males and females and their respective controls. Nevertheless, there was a sex

effect on adrenal weight, with adrenals from male rats weighing significantly less than

those from females. This observation is concordant with data from literature showing that

in wistar rats, sex differences in adrenal weight appear at about PND 49, and from this

day onward, female relative and subsequently absolute adrenal weights are higher than in

male animals (Majchrzak and Malendowicz, 1983).

During the pubertal period, the HPG axis undergoes many dynamic changes

leading to sexual maturation. Puberty marks the final and crucial interactions between

aminergic neurotransmitters systems, gonadal steroids, and hypothalamic-pituitary

hormones (Kalra and Kalra, 1983). Without the coordinated activity of each of HPG axis

components during puberty, alterations in later reproductive functioning are highly

probable (Ojeda et al., 1992).

A number of studies have demonstrated that cocaine modulates the HPG axis. Our

data showed that male rats treated chronically with cocaine throughout adolescence

presented decreased levels of testosterone at 30 minutes after the last administration.

This result agrees with other studies that have previously referred a cocaine effect on

testosterone plasma levels in males. In male rats, acute and binge chronic cocaine

administration are reported to significantly decrease testosterone plasma levels (Berul and

Harclerode, 1989; Budziszewska et al., 1996).

However, the decreased testosterone plasma levels observed at 30 minutes after

the last cocaine administration seems to be the result of its acute effects after chronic

cocaine treatment, since 1 day after withdrawal this alteration was no longer observed. In

contrast to our results, Sarnay et al (1998) reported that in adult male rats, 22 hours after

withdrawal from both 3 and 6 weeks binge cocaine the levels of testosterone were still

decreased. Therefore, the process of maturation taking place during adolescence may

underlie these different results.

140

Chronic cocaine administration during adolescence also did not affect the testes

weight (calculated as percentage of body weight). Despite that, and although the

testosterone plasma levels were not altered in male rats 1 day after the end of chronic

cocaine administration, the repeated decrease in testosterone levels induced by the

“acute” effects of each cocaine administration throughout the exposure period, may have

induced alterations in the developmental process of the reproductive function, which can

lead to long-term impairment in the reproductive function.

The mechanisms of cocaine-induced decrease on the testosterone levels are still

unclear. One of the possible mechanisms underlying the suppression of testosterone

secretion could be the activation of the HPA axis resulting an inhibitory HPA/HPG

interaction (Rivest and Rivier, 1995; Gore et al., 2006). Alternatively, cocaine might act

directly on the testes to decrease testosterone secretion. This suggestion is supported by

morphological changes and lesions of the cells of seminiferous tubules and interstitial

cells of the testes in rats subjected to long-term cocaine administration which may

produce a decrease in testosterone secretion (Barroso-Moguel et al., 1994; George et al.,

1996).

Beside the decreased levels of testosterone on adolescent male rats treated

chronically with cocaine, our data also indicate a trend toward an increase on

progesterone levels at 30 minutes after the last administration. In fact, it has been

demonstrated that after single-, binge- and chronic-cocaine administration paradigms, in

male and female rats, the plasma levels of progesterone were increased (Quinones-Jenab

et al., 2000; Walker et al., 2001; Quinones-Jenab et al., 2008).

The findings that testosterone, but not progesterone plasma levels are reduced

following administration of cocaine suggest that this psychostimulant may interfere with

some specific metabolic processes in the synthesis of testosterone in Leydig cells (figure

3.4.1). Nevertheless, a study in female rats, has shown that cocaine stimulate

corticosterone and progesterone secretion in sham-adrenalectomized, but not in

adrenalectomized rats (Walker et al., 2001), suggesting that the adrenal gland is the

source of cocaine-stimulated progesterone. In this view, the trend to an increase in

progesterone levels may be induced by the increased ACTH levels observed in cocaine-

treated males at this assessment time point. In fact, Feder and Ruf (1969) reported that

systemic injections of ACTH increased plasma progesterone concentrations in

ovariectomized, estrogen-primed rats and guinea pigs (Feder and Ruf, 1969). In the case

of the adrenals being actually the source of progesterone increase, the decreased

testosterone levels, could be explained by the inhibitory effect of the HPA axis on the LH

release (Olster and Ferin, 1987; Petraglia et al., 1987).

141

Figure 3.4.2.1- Pathways of testosterone biosynthesis in the testes. The conversion of pregnenolone into

testosterone requires four enzymatic reactions, which are divided into two parallel pathways: one proceeding

via 17-hydroxypregnenolone, and the other proceeding via 17-hydroxyprogesterone. The relative activities of

the pathways vary among mammalian species; in rodents the pathway via 17-hydroxyprogesterone is

predominant (Norman and Litwack, 1998). 1- P450 side chain cleavage; 2,- 17α-hydroxylase/ C-17-C-20

lyase; 3- 3β-Steroid dehydrogenase; 4- 17-Ketosteroid redutase; 5- 5α-Reductase. Adapted from (Norman

and Litwack, 1998).

Preclinical studies have consistently shown that cocaine stimulates significant

increases in LH levels. Alterations in LH pulsatile release patterns and/or abnormal LH

levels could disrupt gonadotropin and ovarian steroid feedback systems. In fact, cocaine

administration disrupts estrous cyclicity and normal rates of ovulation in rats (King et al.,

1990), and menstrual cyclicity and ovulation rates in monkeys (Potter et al., 1999).

Cocores at al. reported that chronic cocaine abuse in humans was also associated with

menstrual cycle irregularities (Cocores et al., 1986). Nevertheless, the effects of cocaine

on cyclic reproductive function, including circulating LH concentrations, estrous cyclicity

and rats of ovulation in rats are dose-dependent (King et al., 1993).

Our results showed that adolescent female rats chronically exposed to binge

cocaine did not present altered levels of estradiol or progesterone when assessed 30

minutes and 1 day after the last administration. Since in our study, hormonal data from

female rats was analyzed regardless physiological hormonal fluctuations, hormonal

alterations induced by cocaine could have been masked by this approach. Although the

142

effects of cocaine on progesterone levels is known to be affected by the endocrinological

profile of female rats (Quinones-Jenab et al., 2000), several studies that also analyzed the

endocrine profile regardless of the estrous cycle have reported increased levels of

progesterone after cocaine administration (Quinones-Jenab et al., 2000; Quinones-Jenab

et al., 2008). However, cocaine modulation of progesterone plasma levels was transient,

as 3 hours post-injection progesterone plasma levels returned to control levels (Quinones-

Jenab et al., 2000). Based in this observation, it has been suggested that it is possible

that the disruptive effects of cocaine on the HPG axis are transient and/or completely

reversible. Our findings also seem to support the hypothesis that tolerance to female HPG

axis alterations may occur after continued and repetitive use of cocaine.

Moreover, data from literature also shows that cocaine-treated female rats

(20mg/kg/day) from PND 21 to 60, a period that include the pubertal development,

revealed no cocaine effects on the onset of puberty, on first ovulation age and that the

ovulation rates did not differ from the saline controls, once again suggesting that cocaine

did not induced persisting effects on HPG axis in females (Raap et al., 2000).

In what concerns estradiol, it has been reported that estradiol plasma levels are

increased by cocaine in the follicular phase but not in the midluteal phase of the rhesus

monkey cycle (Mello et al., 2000). However, no changes in estradiol levels after acute

cocaine administration have been reported in female rats (Walker et al., 2001).

Nevertheless, the stress induced by the injection protocol may also have lead to

the absence of cocaine effects in progesterone and estradiol plasma levels. It was

reported that footshock stress during 14 days had no effect on estrous cycle length,

estradiol or progesterone concentrations (Anderson et al., 1996). However, in the latter,

like in the present study, animals were housed individually, and a recent report from

Backer and Bielajew (2007) showed that single-housed rats presented greatest cycle

disruptions during post-chronic mild stress than paired housed females, this way

suggesting that housing condition, more than exposure to stress, appeared to contribute

to the disruption of estrous cycling (Baker and Bielajew, 2007). In the present study the

animals were housed individually in order to avoid variable social influences that could

play a role in cocaine behavioural, neurochemical and hormonal effects (de Jong et al.,

2007), however, this approach may have contributed to the lack of differences regarding

female gonadal hormones.

The levels of testosterone were also measured in the adolescent female rats, and

although no differences were observed between cocaine-treated and control females in

the two assessment points, in the cocaine exposed female group the levels of

143

testosterone decreased from 30 minutes to 1 day after the last administration of cocaine.

As testosterone is known to play a role in the reproductive function (Drouin and Labrie,

1976; McClamrock et al., 1991; Weiss et al., 2007), this alteration may be an indication of

a certain degree of cocaine effects on the reproductive function.

During the adolescent developmental period, beside the effects of cocaine on the

HPG axis regulation, the effects of gonadal hormones on the maturating CNS represent

an important issue. Because of the profound effect of gonadal hormones on the

modulation of the CNS plasticity, cocaine-induced alterations in the HPG axis may play a

key role in the modulation of CNS neuronal functions of both males and females and thus

be an important component in the cascade of events that follow the administration of

cocaine (Quinones-Jenab et al., 2001). Thus, the altered levels of testosterone observed

in both male and female rats after chronic cocaine administration throughout adolescence

may have led to disruption of the CNS maturation process, as for instance, testosterone

seems to be implicated in cocaine-induced behavioural sensitization (Martinez-Sanchis et

al., 2002).

3.4.3. Effects of chronic cocaine treatment during adolescence on the dopaminergic and serotonergic systems function

The neural mechanisms responsible for the enhanced adolescent vulnerability for

initiating drug abuse are unclear. Drug exposure during adolescence may have unique

effects because of the dynamic ongoing neural development (Giedd et al., 1999; Spear,

2000; Cunningham et al., 2002; Chambers et al., 2003).

- Effects on the mesoaccumbens and nigrostriatal pathways

The involvement of nigrostriatal and mesolimbic DA in the mediation of

psychostimulant effects is largely supported in literature. The dopaminergic neurons of the

SN and VTA play prominent roles in novelty seeking as well as reinforcement (Le Moal

and Simon, 1991; Spanagel and Weiss, 1999). Although all of the neurochemical

mechanism for dopaminergic transmission is present soon after birth, a number of indices

of dopaminergic function change markedly during adolescence. Overexpression of D1,

D2 and D4 receptors and subsequent decreases during adolescence have been reported

in several studies (Teicher et al., 1995; Tarazi et al., 1998; Montague et al., 1999; Tarazi

et al., 1999; Andersen et al., 2002). Such pruning has also been shown in postmortem

studies of adolescent human brain for DA receptors and DAT (Meng et al., 1999). Thus, to

better understand how addictive drugs affect the DA neurotransmission as dopaminergic

144

systems approach final maturation during adolescence is critical to understanding the

drug abuse vulnerability of this unique developmental period.

Our data showed that 30 minutes after the last administration, adolescent rats

treated chronically with cocaine presented no differences on the levels of DA and its

metabolites, DOPAC and HVA, in the SN-VTA and in the terminal areas, Nac and dorsal

striatum, as compared to saline controls.

Studies using microdialysis have shown that acute cocaine administration leads to

an increase in the extracellular levels of DA (Hurd and Ungerstedt, 1989; Kalivas and

Duffy, 1990). However, after chronic cocaine administration the increase in DA levels is

not so clear. Decreases in basal DA levels in dorsal striatum and Nac have been found in

rats submitted to chronic binge cocaine injection paradigm (Maisonneuve et al., 1995), in

rats injected with cocaine twice daily for 9 or 16 days (Imperato et al., 1992; Rossetti et

al., 1992) and even in rats which received a single daily injection of cocaine for 10 days

(Parsons et al., 1991) or 18 days (Robertson et al., 1991). A suggested mechanism for

this lowered basal level of DA is an increase of the DA reuptake process. For example,

the density of the DAT binding sites in the striatum of rats treated for 14 consecutive days

with cocaine were found to be significantly increased compared to controls after 3 days of

withdrawal (Claye et al., 1995). In a postmortem study of human cocaine-exposed

subjects, DAT binding was increased in the dorsal striatum and Nac (Little et al., 1993).

Thus, an enhanced clearance of synaptic DA may underlie the observed lower levels of

DA. On the other hand, super-sensitivity of D2 DA autoreceptors in the dopaminergic

terminals has been suggested as a mechanism underlying DA decreases (Dwoskin et al.,

1988). Such a change could lead to decreased DA synthesis following chronic cocaine

administration (Trulson and Ulissey, 1987; Brock et al., 1990). However, Tsukada (1996)

reported that cocaine administered to rats for 14 days in a binge pattern produced a

significant reduction in the in vivo D1 and D2 binding in striatum, due mainly to alterations

in the affinity (Tsukada et al., 1996). Nevertheless, Koeltzow and White (2003) reported

that binge cocaine-treated rats exhibited a decreased in the number of spontaneously

active VTA DA neurons. Another suggested mechanism for the lowered basal level of DA,

is that alterations in the expression of neuropeptides that modulate concentration of DA

that have found to play an important role. For example, increases in the levels of mRNA of

the striatal neuropeptide dynorphin have been found following chronic administration of

cocaine to rats (Sivam, 1989; Spangler et al., 1993; Daunais and McGinty, 1995).

Dynorphin is known to lower the striatal DA release (Reid et al., 1988). More recently, it

was reported that prolonged treatment with cocaine reduces dopaminergic terminal

density in regions receiving projections, and this reduced dopaminergic terminal density

145

that follows prolonged treatment with cocaine was associated with reduced basal levels of

DA (Horne et al., 2008).

Nevertheless, and as already mentioned, our data showed no differences in levels

of DA and its metabolites in the SN-VTA, Nac and dorsal striatum, 30 minutes after the

last administration, between cocaine-treated and control male and female adolescent rats.

This absence of differences may be related to the lower basal levels of DA reported to

occur in chronically treated animals, and may not represent an effective absence of

increase in levels of DA induced by the binge cocaine administration. However, these

results contrast with previous studies that reported small increases on DA levels in the

striatum after cocaine binge during chronic exposure (Maisonneuve et al., 1995; Zhang et

al., 2003).

The immaturity of the dopaminergic neurotransmission during adolescence may

underlie the different response to the binge administration. In fact, adolescent and adult

rats use a different balance of mechanisms to attain comparable rates of DA clearance.

Relatively to adults, adolescents present a higher ratio of uptake/release, and this uptake-

dominated situation likely leads to lower extracellular DA levels during adolescence

(Walker and Kuhn, 2008). This developmental specific type of regulation could influence

drug-induced activation of the dopaminergic system (Walker and Kuhn, 2008).

In fact, basal extracellular DA concentration measured by microdialysis are

reported to be lower in adolescent than in adult rats (Andersen and Gazzara, 1993;

Laviola et al., 2001). In contrast to lower extracellular DA in adolescents, striatal D1 and

D2 DA receptor levels are higher in adolescent than adult humans (Palacios et al., 1988;

Montague et al., 1999) and rats (Tarazi et al., 1998, 1999), and DAT density reaches

maximal expression during adolescence (Haycock et al., 2003). The combination of an

increased uptake and a decreased synthesis due to the higher D2 receptor levels, could

be the reason for the lower increase in levels of DA observes in this study after a binge

administration.

The mechanism of how a hypofunctional dopaminergic system would produced

greater vulnerability to addiction is not fully understood (Walker and Kuhn, 2008). One

possibility is that the adolescent animals have lower baseline dopaminergic activity and an

enhanced threshold for rewarding stimuli (Bjork et al., 2004).

Nevertheless, it has been hypothesized that decreases in levels of DA in the brain

are associated with the long-lasting dysphoric state and craving after cocaine withdrawal

reported in humans abstinent from cocaine (Dackis and Gold, 1985). Such a change may

play a significant role in maintaining or reinstating drug-taking to compensate for

146

decreased levels of DA during withdrawal (Dackis and Gold, 1985; Weiss and Porrino,

2002).

Based on data obtained from chronic repeated cocaine-induced alterations on the

dopaminergic system, and on the age-specific characteristics of dopaminergic function of

adolescents it was expected that 1 day after withdrawal, the levels of DA in the

mesoaccumbens and nigrostriatal systems would be decreased. Our data showed that

cocaine-treated male animals did not display any difference on DA or its metabolites,

DOPAC and HVA levels in Nac as compared to control males. However, in the dorsal

striatum, DA turnover, calculated as (DOPAC+HVA)/DA was increased in cocaine-treated

males, and in the SN-VTA the levels of DA and DOPAC were increased.

Since animals were sacrificed in the room where cocaine was administrated, we

suggest that this absence of lower levels of DA could be related with the cocaine cues-

induced increases in DA. In fact, several investigators have demonstrated stimulant-like

effects when drug-free rats are exposed to environments that have been previously paired

with stimulant drugs, such as amphetamine or cocaine (Post et al., 1981; Badiani et al.,

1995). Moreover, it was shown that conditioned rewarding stimulus associated with

cocaine self-administration reverses the depression of catecholamine brain systems

following cocaine withdrawal in rats (Antkiewicz-Michaluk et al., 2006).

Dopamine, which is a neurotransmitter involved with reward and with the prediction

of reward (Wise and Rompre, 1989; Schultz et al., 1997), appears to be involved with cue-

elicited drug craving. Indeed, in laboratory animals when neutral stimuli are paired with a

rewarding drug they will, with repeated associations, acquire the ability to increase DA in

Nac and dorsal striatum, and these neurochemical responses are associated with drug-

seeking behaviour (Kiyatkin and Stein, 1996; Duvauchelle et al., 2000; Weiss et al., 2000;

Phillips et al., 2003; Di Ciano and Everitt, 2004; Vanderschuren et al., 2005). Imaging

studies in humans have also shown that in drug-addicted subjects visual exposure to drug

cues elicits DA increases in dorsal striatum and these increases are associated with the

subjective experiences of drug craving (Volkow et al., 2006; Wong et al., 2006).

Thus, during a drug-free period, on the day after cocaine administration an

increase in the levels of DA in the SN-VTA and the absence of alterations in DA and its

metabolites in Nac in rats previously treated with cocaine may be a consequence of the

exposure to stimuli associated to cocaine administration. Nevertheless, a sex difference in

DOPAC levels 30 minutes after the last administration was observed in the SN-VTA in

cocaine-treated animals.

147

In fact, adolescent rats have been reported to be more sensitive to the place

conditioning effects of cocaine, showing preferences after conditioning with lower doses

than adults (Badanich et al., 2006). Clinical research suggests that adolescence is indeed

a critical period when motivational conditioning is heightened (Harrison et al., 1989).

Actually, a hypothesis was developed that defends that adolescent vulnerability to

addiction involves a developmental state that is primed to assign high salience to reward-

related cues (Brenhouse and Andersen, 2008).

The reinforcing properties of cocaine are thought to be due to the ability of cocaine

to block the uptake of DA. However, mapping of the sites of cocaine binding and neuronal

activation in DAT knockout mice suggests the involvement of serotonergic brain regions in

initiation and maintenance of cocaine self-administration (Rocha et al., 1998). Indeed,

cocaine self-administration in DAT knockout mice suggests that the serotonergic system

may provide an additional component of drug reinforcement (Rocha et al., 1998).

It is known that cocaine also inhibits the reuptake of 5-HT into serotonergic

neurons (Reith et al., 1983; Pitts and Marwah, 1987), and this effect of cocaine leads to

elevation of extracellular 5-HT levels in the brain (Bradberry et al., 1993; Parsons and

Justice, 1993a; Reith et al., 1997). Increased extracellular 5-HT levels in striatum and VTA

after acute cocaine have been reported (Bradberry et al., 1993; Reith et al., 1997; Muller

et al., 2002). After chronic treatment (20 mg/kg i.p. for 10 consecutive days) a systemic

injection of cocaine also induces higher increases of 5-HT levels in the Nac and VTA as

compared to saline control rats (Parsons and Justice, 1993a). These results contrast with

those obtained in our study. We did not observe any differences in 5-HT and 5-HIAA

levels between cocaine-treated and control animals 30 minutes after the last binge

treatment. More recently, Mills at al. (2007) reported that in rats, chronic cocaine

administration (20 mg/kg/day for 18 days) did not increase 5-HT levels in the caudate

nucleus, although produced significant increases in the Nac (Mills et al., 2007).

Some evidence supports the notion that withdrawal after repeated cocaine

administration in rats may leads to diminished synaptic 5-HT availability (Parsons et al.,

1995). Alterations in 5-HT transport may contribute to the post cocaine deficits in

serotonergic neurotransmission. Increased densities of 5-HT reuptake sites have been

demonstrated in rats following repeated cocaine exposure (Cunningham et al., 1992). In

studies using single photon emission computed tomography, acutely abstinent cocaine-

dependent patients had higher SERT binding in striatal brain regions compared to non-

drug abusing control subjects (Jacobsen et al., 2000). In monkeys, self-administration of

cocaine also induced significantly higher levels of SERT availability in the dorsal striatum

compared to control subjects (Banks et al., 2008). In fact, it has been suggested that

148

chronic cocaine-produced alterations in the serotonergic neurotransmission may underlie

the dysphoria experienced by abstinent cocaine users leading to maintenance of drug

abuse (Baumann et al., 1995; Ghitza et al., 2007).

Our data also did not showed any differences in 5-HT levels between cocaine-

treated and control animals 1 day after cocaine withdrawal in Nac and dorsal striatum.

These results contrast with data from self-administration studies that reported decreased

5-HT in the Nac during withdrawal from cocaine self-administration (Parsons et al., 1996).

However, in accordance with our results, studies with passive administration of cocaine

also reported unchanged striatal levels of 5-HT, 1 day after 14 days of repeated

administration of cocaine (20 mg/kg/dose, i.p. every 12 h) (Johnson et al., 1993). The

experimental method in this study seems to approach more to our experimental

conditions, leading to similar results. Thus, the fact that cocaine has been passively

administrated may have contributed to the absence of differences between cocaine-

treated and control animals.

Nevertheless, our data also showed increased levels of 5-HT in the SN-VTA, 1 day

after the last administration in males chronically treated with cocaine.

The existence of a functional relationship between 5-HT and DA neurons in the

mesolimbic dopaminergic system has been confirmed (McMahon et al., 2001). Serotonin

receptor localization in mesolimbic circuits supports the possibility that 5-HT modulates

the flow of information through dopaminergic mesolimbic circuits, and thus these 5-HT/DA

interactions may participate in the elicitation of cocaine effects.

Neuroanatomical, electrophysiological and biochemical evidence is provided for an

interaction between serotonergic and dopaminergic systems in different brain regions,

including the SN-VTA. Dorsal raphe stimulation or local application of 5-HT agonists and

antagonists to the SN or the VTA, modulates the electrophysiological activity of

dopaminergic neurons (Kelland et al., 1990; Trent and Tepper, 1991; Pessia et al., 1994;

Prisco et al., 1994; Brodie and Bunney, 1996; Minabe et al., 1996; Cameron et al., 1997).

The 5-HT-induced release of DA has been demonstrated in vitro, in striatal (Blandina et

al., 1989) and nigral slices (Williams and Davies, 1983) and in vivo, using microdialysis in

the Nac (Parsons and Justice, 1993b; Hallbus et al., 1997), striatum (Benloucif and

Galloway, 1991), SN (Thorre et al., 1998) and in the prefrontal cortex (Iyer and Bradberry,

1996). Based on this, the observed increased levels of 5-HT in SN-VTA in male rats may

be contributing to the higher dopaminergic response to cocaine-cues observed in male

rats, this way leading to the enhanced adolescent vulnerability to addiction.

149

- Effects on the amygdala

Modulation of neuronal plasticity in the mesoaccumbens dopaminergic system,

which is thought to be essential in the mediation of the behavioural effects of drugs of

abuse (Koob et al., 1998; Di Chiara et al., 2004), has been demonstrated in a number of

studies (Wolf et al., 2004; Jones and Bonci, 2005). However, psychostimulant dugs also

influence plasticity in memory-related brain regions such as the hippocampus (Thompson

et al., 2002; Thompson et al., 2005) and the amygdala (Goussakov et al., 2006).

The amygdala is an important component of prefrontal cortex networks by its

involvement in decision-making processes as it encodes the emotional value/intensity of

environmental stimuli and outcome–action associations (Winston et al., 2005; De Martino

et al., 2006). Basolateral amygdala neurons relay this information by excitatory efferents to

pyramidal neurons in the medial prefrontal cortex (Krettek and Price, 1977; Gabbott et al.,

2006; Orozco-Cabal et al., 2006). However, after chronic cocaine use, basolateral

amygdala/ medial prefrontal cortex neurotransmission may become aberrant as cocaine-

dependent individuals exhibit inappropriate utilization of emotional information to guide

behaviour (Schoenbaum et al., 2006).

Among the various neurotransmitter inputs to the amygdala, several lines of

evidence have implicated dopaminergic inputs from the VTA as having a crucial

importance (Brinley-Reed and McDonald, 1999). Amygdala also expresses both the D1

and D2 receptor (Meador-Woodruff et al., 1991; Scibilia et al., 1992). Within the

amygdala, DA stimulates D1 and D2 receptors post-synaptically on glutamatergic

projection neurons (Rosenkranz and Grace, 2002) and GABAergic interneurons (Bissiere

et al., 2003), as well as pre-synaptically on D2 DA autoreceptors (Bull et al., 1991).

In the present study no differences were observed in levels of DA or its metabolites

in amygdala 30 minutes after the last cocaine administration in both males and females as

compared with controls. Wilson et al. (1994) reported that DA and 5-HT levels, 1 h after

the last cocaine session, were significantly elevated in amygdala of rats self-administering

cocaine, but not in rats passively injected, provide additional support for the involvement

of the amygdala in the acquisition of drug seeking behaviour associated with cocaine self-

administration.

One day after the end of cocaine administration the levels of DA were not different

between cocaine-treated males and controls in the amygdala. However, cocaine-treated

adolescent females, as compared to control females, displayed increased levels of DA in

the amygdala 1 day after the last administration of cocaine.

150

It was already reported that the environmental stimuli predictive of cocaine

availability elevate extracellular DA levels in the amygdala (Weiss et al., 2000).

Suggesting that increased DA transmission in this brain region may be one of the

neurobiological mechanisms underlying cocaine craving associated with exposure to

drug-related stimuli. Moreover, rats systemically administered D1 DA receptor

antagonists, reduce cocaine-seeking behaviour and Fos expression in the basolateral

amygdala in the presence of a cocaine-predictive stimulus (Ciccocioppo et al., 2001).

Additionally, intra basolateral amygdala infusion of D1 receptor antagonist abolishes the

expression of conditioned-cued reinstatement of cocaine-seeking behaviour (See et al.,

2001). While basolateral amygdala infusion of amphetamine potentiates it (Ledford et al.,

2003). Evidence from functional magnetic resonance imaging and positron-emission

tomography studies in humans also showed that cue-induced cocaine craving is

associated with activation of DA-rich forebrain regions including the amygdala (Grant et

al., 1996; Childress et al., 1999). Thus, the increased levels of DA displayed by females

may be related with the exposure, at the time of decapitation, to an environment stimulus

paired with cocaine administration. Our results seem to point to a sex-effect on the

response to cocaine-related stimuli 1 day after the last administration of cocaine, with

females showing a greater involvement of the amygdala.

In this study the levels of 5-HT in the amygdala were also measured 30 minutes

and 1 day after the last administration of cocaine. No differences were observed between

cocaine-treated and controls animals, both in male and female adolescent rats. As already

mentioned, Wilson et al. (1994) reported that when compared to the controls, in rats self-

administering cocaine, 5-HT levels 1 h after the last cocaine session, were significantly

elevated in amygdala of rats self-administering cocaine, but not of rats passively exposed

to cocaine.

- Effects on the prefrontal cortex

It is known that acute cocaine dose-dependently increase DA in the prefrontal

cortex (Pum et al., 2007). However, repeated cocaine administration has been shown to

attenuate the increase in prefrontal cortex extracellular DA levels elicited by a systemic

cocaine injection (Sorg et al., 1997; Chefer et al., 2000; Williams and Steketee, 2005b).

Our results did not show any difference in DA or its metabolites levels 30 minutes

after a binge administration in the prefrontal cortex. This could be related with the reported

attenuated increases in extracellular DA after chronic treatment. Like in our study, others

have found that the DA response to an acute cocaine injection 1 day after a repeated

151

cocaine treatment (cocaine 4x15mg/kg/day i.p. for 4 days) was blunted (Williams and

Steketee, 2005b). Moreover, it has been reported that repeated cocaine produced no

statistically significant changes in baseline extracellular DA levels within the prefrontal

cortex (Sorg et al., 1997; Chefer et al., 2000; Williams and Steketee, 2005b, a). Based on

this, the lower increases in DA were probably the result of cocaine-mediated reductions in

the release of DA. It was demonstrated that this is not associated with significant changes

in DA clearance or DAT protein, but may be related with an hyporesponsivity in

mesocortical neurons mediated by a transient attenuation in depolarization-induced DA

release (Williams and Steketee, 2005a).

It was first proposed that a potential mechanism underlying the attenuation in

cocaine-induced DA overflow in the prefrontal cortex could be that repeated exposure to

cocaine would enhance DA clearance within prefrontal cortex nerve terminals. However a

recent study (although in contrast to previous findings for the Nac and dorsal striatum)

showed that repeated cocaine was not associated with significant changes of DAT protein

expression in either whole synaptosomes or the plasma membranes of the medial

prefrontal cortex (Williams and Steketee, 2005a). It appears possible that other effects of

cocaine may be taking place presynaptically in the medial prefrontal cortex at the level of

the dopaminergic nerve terminals, including cocaine-mediated changes in the DA

synthesis, vesicular storage, or autoregulation of release, which have been shown to be

altered in other mesocorticolimbic and mesostriatal brain regions after repeated

administration of psychostimulants (Trulson and Ulissey, 1987; Beitner-Johnson and

Nestler, 1991; Brown et al., 2002). Thus, the low cocaine-induced increases in DA levels

may result from a decreased DA releasability rather than an enhanced DA clearance

(Williams and Steketee, 2005a).

Within the prefrontal cortex, DA released from mesocortical neurons generally

attenuates the activity of pyramidal glutamatergic neurons, the primary excitatory efferents

of the prefrontal cortex, either by its direct effect on these cells or indirectly through

stimulation of local aminobutyric acid (GABA) release from local inhibitory interneurons

(Sesack and Bunney, 1989; Cowan et al., 1994; Grobin and Deutch, 1998). Based on the

cellular and neuroanatomical arrangement of the prefrontal cortex, attenuation in cocaine-

induce increase in dopaminergic transmission in the prefrontal cortex, after repeated

cocaine exposure, could produce a disinhibition of the prefrontal cortex pyramidal neurons

resulting in an increased excitatory output to subcortical regions. Enhanced excitatory

transmission in the medial prefrontal cortex has previously been associated with

neuroadaptive processes, within the VTA, involved in the induction of sensitization (Wolf

et al., 1995; Li et al., 1999). Furthermore, prefrontal glutamate release to the Nac has

152

been shown to be critical for the expression of sensitization (Pierce et al., 1996; Pierce et

al., 1998) and the reinstatement of cocaine self-administration (McFarland et al., 2003).

In our study, 1 day after the last administration of cocaine the levels of DA in

cocaine-treated animals did not differ from controls, this is in agreement with other studies

already mentioned, that reported no significant changes in baseline extracellular DA levels

within the prefrontal cortex after repeated cocaine administration (Sorg et al., 1997;

Chefer et al., 2000; Williams and Steketee, 2005b, a).

It is also known that acute cocaine dose-dependently increases 5-HT in the

prefrontal cortex (Pum et al., 2007). However, it has been reported that chronic cocaine

does not alter the basal levels of cortical 5-HT (Baumann et al., 1993; Johnson et al.,

1993). However, it causes regionally specific alterations in 5-HT uptake binding

(Cunningham and Callahan, 1991). In the prefrontal cortex it was showed a cocaine-

induced elevation in 5-HT uptake sites (Cunningham et al., 1996). Recently, it was

reported that prolonged cocaine administration increases SERT terminal density in the

cortex and reduced 5-HT1A autoreceptors binding on serotonergic neurons projecting

from raphe, suggesting that for serotonergic neurons the autoreceptor feedback would

likely diminish following long-term treatment with cocaine (Horne et al., 2008). This

mechanism is likely to contribute towards the sprouting of serotonergic terminals. Thus,

the absence of differences between cocaine- and saline-treated animals, both 30 minutes

and 1 day after the last administration, observed in our study, may be related with

cocaine-induced adaptive processes in the serotonergic function within this area.

Prefrontal cortex serotonergic system plays a pivotal role in the locomotor

stimulant effect of cocaine (Nakamura et al., 2006). A role of the medial prefrontal cortex

in the mediation of the conditioned preference for environments associated with

psychostimulant drugs it has also been demonstrated. Excitotoxic lesions of the medial

prefrontal cortex blocked the acquisition of conditioned place preference (Tzschentke and

Schmidt, 1999). Pum et al. (2008) went further by showing that conditioned place

preference may be mediated, at least in part, by the serotonergic innervation of the medial

prefrontal cortex. Depletion of 5-HT in the medial prefrontal cortex significantly attenuated

the preference for the cocaine-associated environment and the hyperlocomotor response

to cocaine (Pum et al., 2008).

153

- Effects on the hippocampus

Like observed for the prefrontal cortex, in the hippocampus no differences were

found on levels of DA or its metabolites both in males and female rats chronically exposed

to cocaine, both 30 minutes and 1 day after the last administration of cocaine. The

hippocampus is an important mediator of learning and reinforcement, but its role in

cocaine effects has received little attention.

The role of DA in the hippocampus has not been extensively studied because of

the early view that the hippocampus did not receive a significant dopaminergic innervation

(Loy et al., 1980). However, it is now clear that the hippocampus does receive such

innervation (Gasbarri et al., 1994) and there has been progress in understanding its

function. Specifically, work in the field of synaptic plasticity has provided clear evidence

that DA affects long-term potentiation (LTP), a form of synaptic plasticity thought to

encode long-term memory. The hippocampus receives dopaminergic inputs which arrive

both from the SN and the VTA (Scatton et al., 1980). Thus, cocaine-induced alterations in

the dopaminergic function may influence the hippocampus function.

It was expected that cocaine would enhances the levels of DA in hippocampus,

since Thompson et al. (2005) reported that cocaine can enhanced LTP magnitude in a

hippocampal slice assay, and that this modulation of LTP is probably a result of cocaine’s

blockade of DAT that promotes enhanced catecholamine transmitter levels, leading to the

activations of D2-like receptors. The D2 DA receptor subtype has previously been

characterized as having positive effects on LTP (Frey et al., 1990).

However, in the present study, the levels of DA were not increased 30 minutes

after the last administration of cocaine, which may be related with adaptations to the

chronic cocaine exposure already mentioned for other brain regions, such as the reduced

density of dopaminergic terminals reported by Horne at al. (2008) for the striatum (Horne

et al., 2008), or a decrease in cocaine-induced raise in DA levels in result of a decreased

DA releasability (as proposed for prefrontal cortex (Williams and Steketee, 2005a)).

Nevertheless alterations on the hippocampal function after cocaine exposure may

contribute to the maintenance of drug use. Several findings indicate that the ventral

hippocampus plays an important role in the relapse to cocaine-seeking behaviour (Rogers

and See, 2007). Selective inhibition of the ventral hippocampus attenuates relapse to

cocaine seeking following presentation of drug-paired cues or a priming injection of

cocaine itself (Rogers and See, 2007). One obvious source of regulation is the

dopaminergic input to ventral hippocampus. Lisman and Grace (2005) have recently

suggested that DA critically gates the amount of information processed by the CA1

154

subregion of the hippocampus. However, the hippocampus is also involved in the

modulation of other DA-innervated regions implicated in cocaine addiction (Rogers and

See, 2007). In fact, stimulation of the ventral subiculum of the hippocampal formation

induces long-lasting DA release in the NAc (Brudzynski and Gibson, 1997; Legault et al.,

2000) and enhances the firing of mesolimbic DA neurons that originate in the VTA (Todd

and Grace, 1999; Legault et al., 2000). This Nac DA increase after ventral subiculum

stimulation depends on VTA glutamate. In fact, Nac DA increase is blocked by the

nonselective ionotropic glutamate receptor antagonist, kynurenic acid, when applied into

the VTA (Legault et al., 2000). Increased impulse flow through VTA DA neurons mediated

by excitatory glutamate inputs to this region, appears critical for ventral subiculum

stimulation to elevate Nac DA (Legault et al., 2000).

The substantial serotonergic innervation of the hippocampus (Jacobs and Azmitia,

1992), which is known to regulated hippocampal activity (Azmitia et al., 1984), raises the

possibility that cocaine also influences serotonergic transmission in the hippocampus. In

the present study, no differences were found in 5-HT as well as in 5-HIAA levels, 30

minutes after the last administration of cocaine, in both male and female adolescent rats.

In contrast, it is accepted that cocaine increases the extracellular 5-HT concentration in

the hippocampus when administrate acutely (Muller et al., 2002; Muller et al., 2004).

However, the chronic effects on 5-HT concentration are mostly unknown. Probably the

absence of differences between cocaine-treated and control animals reflects the

development of adaptation processes.

The activity of the principal cells in the hippocampus is under control of GABAergic

interneurons (Freund and Antal, 1988; Freund et al., 1990). The cocaine-induced increase

in extracellular 5-HT concentration is likely to cause a strong activation of hippocampal 5-

HT1A receptors and thus an inhibition of the GABAergic interneurons (Gulyas et al.,

1999). A reduced GABAergic inhibition of the principal cells of the hippocampus increases

activity in this region (Gulyas et al., 1999), leading to the activation of Nac neurons (Yang

and Mogenson, 1984) and DA release in the Nac (Legault et al., 2000).

Cocaine-induced alterations in the activity of hippocampus, through the interaction

with Nac and VTA, will probably contribute to the cocaine reward effects and to the

addiction state.

155

- Effects on the dorsal raphe nucleus

The dorsal raphe nucleus is a major source of 5-HT in the mammalian brain

(Jacobs and Azmitia, 1992). A previous study has bound that in the raphe, under basal

conditions, 5-HT release was proportional to 5-HT recapture, leading to constant

extracellular pools of 5-HT (Adell et al., 1991). As the raphe is poor in 5-HT nerve

terminals, but sensitive to SERT blockade, it was suggested that SERT should be present

in 5-HT soma and dendrites (Adell et al., 1991). Cocaine action on blocking SERT

disrupts this equilibrium between intra- and extracellular levels of 5-HT.

In the present study, data from dorsal raphe nucleus showed that, 30 minutes after

the last administration of cocaine, males presented decreased 5-HT turnover. This may

indicate an acute effect of cocaine blocking SERT, preventing by this way the 5-HT

uptake, lowering the intracellular concentration of 5-HT and therefore reducing the levels

of 5-HT available for metabolization. In cocaine-treated females, the levels of 5-HT

decreased from 30 minutes to 1 day after the last administration, moreover at 30 minutes

these animals presented higher levels of 5-HT that almost reached significance as

compared to controls (p<0.055), this may also be explained by the acute effects of

cocaine in inhibiting the 5-HT reuptake, decreasing the 5-HT metabolization into 5-HIAA.

In fact, in cocaine exposed females the 5-HT turnover increased from 30 minutes to 1 day

after the last administration. Our data, seems to indicate that both males and females

treated with cocaine, presented extracellular increased levels of 5-HT after the last

administration of cocaine. Repeated cocaine administration has been shown to induce

increases of both 5-HT and DA in the dorsal raphe nucleus after an i.p. injection (Parsons

and Justice, 1993a). Based in this data, the absence of cocaine-induced increases in 5-

HT levels in the other analyzed brain regions seems to be mostly related with processes

occurring at terminal areas, and not mainly dependent on 5-HT synthesis area.

5-HT neurons of the dorsal raphe receive a dense dopaminergic innervation from

midbrain DA neurons (Peyron et al., 1995; Kitahama et al., 2000) and express D2-like DA

receptors (D2, D3) (Mansour et al., 1990; Suzuki et al., 1998). These anatomical features

suggest that the DA input to the dorsal raphe may play a critical role in the regulation of

the function of dorsal raphe serotonergic neurons. Thus, alterations on dopaminergic

function induced by repeated cocaine administration could interfere with serotonergic

function on dorsal raphe nucleus. Consistent with this notion, in vivo neurochemical

studies have reported that administration of DA receptor agonists increases the synthesis

and release of 5-HT in the dorsal raphe nucleus (Ferre et al., 1994; Matsumoto et al.,

1996). Direct evidence for a role of DA in the regulation of serotonergic neurons function

comes from a recent in vitro electrophysiological study showing that activation of DA

156

receptors induces a membrane depolarization and thereby increases the excitability of

dorsal raphe nucleus serotonergic neurons (Haj-Dahmane, 2001). This excitatory effect is

mediated via the activation of DA D2-like receptors located on dorsal raphe nucleus 5-HT

neurons. In contrast to the midbrain DA neurons, activation of D2-like DA receptors in the

dorsal raphe nucleus strongly depolarizes and enhances the excitability of 5-HT neurons

(Haj-Dahmane, 2001). Our results did not showed any effect on the levels of DA our its

metabolites, both at 30 minutes and 1 day after the last administration of cocaine, both in

male and female adolescent rats.

- Effects on the cerebellum

Only a few studies have documented the effects of cocaine in the cerebellum.

Functional brain-imaging studies in cocaine-dependent subjects noted impairments in the

cerebellum (Gottschalk and Kosten, 2002), and an animal study has reported cocaine-

induced microscopic lesions in cerebellum (Barroso-Moguel et al., 2002). Through

development in a rat model, we have previously reported altered serotonergic turnover in

the cerebellum (Summavielle et al., 2004). Moreover, the cerebellum is activated after

exposure to cues eliciting cocaine craving (Grant et al., 1996; Wang et al., 1999).

Cerebellar dysfunction may result in neuropsychological deficits as well as impairments in

fine motor control that have been observed in cocaine abusers (Smelson et al., 1999;

Gottwald et al., 2004; Hester and Garavan, 2004).

In the present study the levels of DA and its metabolites, DOPAC and HVA, were

also analyzed in cerebellum, a brain region with low concentration of DAT (Glaser et al.,

2006). We have observed only for exposed males a decrease in DA and HVA levels

between 30 minutes and 1 day after administration, this may result for the acute effects of

cocaine of increase the extracellular levels of DA.

Although rat cerebellum is known to receive dense serotonergic projections

(Takeuchi et al., 1982; Bishop and Ho, 1985), our data did not revealed alteration on 5-HT

levels in cocaine-treated males. However, we have previously reported that neonatal

cocaine exposure affected the serotonergic cerebellar system by increasing its 5-HT

content (Summavielle et al., 2004). Repeated cocaine has been shown to increase the

expression of 5-HT3 receptor in cerebellum, which may be a consequence of an abnormal

serotonergic system (Arpin-Bott et al., 2006). Cocaine-induced increase in 5-HT in

cerebellum may be involved in counterbalancing cell damage that occurs in response to

cocaine administration.

157

- Effects on the hypothalamus

Within the hypothalamus our data also showed that the 5-HT turnover rate was

decreased in cocaine-treated males 30 minutes after the last administration of cocaine,

probably due to the acute effect of cocaine, inhibiting the 5-HT reuptake, decreasing its

metabolization, and leading to increased extracellular levels of 5-HT. In fact, this animals

presented a rise in the levels of 5-HIAA from 30 minutes to 1 day after the last

administration. Moreover, 1 day after the last administration of cocaine the 5-HT turnover

rate was no longer altered. In females, this effect was not observed.

It is known that 5-HT has stimulatory effects on CRH induction in hypothalamus

(Buckingham and Hodges, 1979; Hanley and Van de Kar, 2003; Mikkelsen et al., 2004).

Thus, probably the altered HPA axis function observed 30 minutes after the last cocaine

administration in male rats, as also indicated by the increased plasma levels of ACTH,

was related with an alteration in serotonergic neurotransmission in the hypothalamus.

The HPA axis activation by either acute or chronic “binge” cocaine is apparently

mediated through the DA receptors (Borowsky and Kuhn, 1991; Spangler et al., 1997;

Zhou et al., 2001; Zhou et al., 2004). Cocaine-induced CRH mRNA increase was absent

in rats pretreated with either a selective D1-like or D2-like DA receptor antagonist,

suggesting that cocaine-induced stimulation of hypothalamic CRH gene expression is

secondary to changes in the activity of specific components of dopaminergic systems

(Zhou et al., 2004). In our study the levels of DA and its metabolites, when measured in

hypothalamus, did not reveal any significant differences between cocaine-treated and

control animals.

- Conclusion

In conclusion, the reduced number of alterations on dopaminergic function verified

30 minutes after the last administration of cocaine seems to indicate the development of

tolerance to the acute effects of cocaine. These results follow the majority studies on

chronic binge cocaine administration that report, only slight increases in extracellular

levels of DA after cocaine administration. The immaturity of the dopaminergic system in

the adolescents, and therefore increased plasticity, may explain why in this model there

are even less short-term alterations than in the adult.

To the development of tolerance, 1 day after the end of the administration another

factor must be taken into account to explain the lack of changes on dopaminergic system

in the most areas analyzed and the increase in the dopaminergic activity in SN-VTA and

in the dorsal striatum in males and in amigdala in female rats. In fact, the presentation of a

158

cocaine related stimuli, may mask the expected decrease in dopaminergic

neurotransmission, justifying the absence of more pronounced differences.

Likewise, the serotonergic neurotransmission, 30 minutes after the last cocaine

administration presented only increased 5-HT in the dorsal raphe nucleus, suggesting that

the absence of cocaine-induced alterations in serotonergic neurotransmission in the other

brain regions was related with adaptive process occurring at terminal areas. Additionally,

we observed decreased 5-HT turnover in the hypothalamus, which may be related with

the increased HPA axis activity observed in cocaine-treated male rats. At 1 day after the

last administration the levels of 5-HT were increased in SN-VTA, this effect may had

contribute to the high dopaminergic activity observed in this area, at this assessment time

point.

3.4.4. Effects of chronic cocaine treatment during adolescence on the anxiogenic-like behaviour

The abrupt discontinuation of cocaine use prompts the expression of various

withdrawal symptoms, including depression, fatigue, parasomnias, anhedonia, anxiety,

and craving (Gawin and Kleber, 1986; Weddington et al., 1990; Satel et al., 1991).

Previous works carried out during adulthood have shown that withdrawal from

cocaine administration induces anxiety-like behaviour in rodents (Basso et al., 1999;

Paine et al., 2002). Our data failed to demonstrate anxiety-like behaviour after chronic

cocaine administration during adolescence, through classic EPM measured 2 days after

the last administration of cocaine. However, we have observed, in treated males,

unexpected risk-like behaviour that seems to indicate loss of risk-assessment.

The number of open arm entries and the time spent in open arms, taken as an

index of anxiety, were not affected by the cocaine treatment. Other authors have shown

that chronic cocaine administration itself failed to induce anxiety in the EPM, however

during the withdrawal period, anxiety-like behaviour was observed through decreased

time spent in open arms and decreased number of entries into the open arms were 48 h

after cocaine treatment (20 mg/ kg cocaine i.p., once a day for 14 days in rats) (Sarnyai et

al., 1995). Also, using a different paradigm, conditioned defensive burying, it was

demonstrated that severe anxiety developed within 48 h after the last cocaine injection (20

mg/kg/day, 14 days) (Harris and Aston-Jones, 1993).

Our work was conducted using adolescent rats, and several studies have shown

that cocaine induces different effects on cocaine-related behaviour across ages (Collins

and Izenwasser, 2002; Kerstetter and Kantak, 2007). In contrast to the adults,

159

periadolescent rats did not show behavioural sensitization to cocaine (Collins and

Izenwasser, 2002). A similar response was observed in amphetamine exposed rats, which

showed diminished behavioural responsiveness to amphetamine during periadolescence

(PND 30-40) (Bolanos et al., 1998). Moreover, adolescent animals are known to have a

unique behavioural profile even in response to an acute administration of

psychostimulants. A profile of hyporesponsitivity to locomotor effects of an acute

administration of psycostimulants has been reported (Spear and Brake, 1983; Adriani and

Laviola, 2000). Adolescent rodents markedly differ from adults in their spontaneous

behaviour (Laviola et al., 1999; Spear, 2000), and this also could be the reason for the

lack of anxiety-like response after cocaine withdrawal observed in our results.

Additionally, aging rats prenatally exposed to either high-dose cocaine

(40mg/kg/day) or high-dose nicotine (5.0 mg/kg/day) were less timid/more impulsive,

spending most of the time in the open arms and having an high percentage of entries into

the open arms of the EPM (Sobrian et al., 2003). Moreover, in a recent work in which

adolescence mice were used, it was shown that the group-housed mice treated with a

binge pattern of cocaine were, when adults (2 weeks after the last administration), less

anxious than those treated only with saline. Interesting, such effect was not present in the

corresponding adult-treated animals (Estelles et al., 2007). All together these data

reinforces the notion that the timing of exposure strongly affects the cocaine-induced

anxiety.

Importantly, in our study, cocaine-treated male rats performed behaviours like fell-

or jump-off the apparatus and walk on the top of the closed arms during the EPM. These

unexpected behaviours were assembled in one category that we call risk-like behaviour.

This happened with an elevated frequency, six out of nine animals showed these

behaviours, and is strengthened by the fact that neither females nor male saline controls

expressed any type of risk-like behaviour. On the other hand, cocaine exposed male rats

also showed a decrease in the time spent in the central platform and a trend towards

decreased number of central platform entries. The behavioural repertoire of cocaine-

treated male rats observed during acute withdrawal is likely to be a consequence of a less

cautious behaviour while exploring the novel environment provided by the EPM

apparatus. Moreover, the risk-like behaviour was independent of any change in locomotor

activity, as there were no differences in the number of the closed arm entries, the most

reliable measure of locomotor activity on the maze (Hogg, 1996). This lack of

disturbances in motor activity during cocaine withdrawal is consistent with data obtained

from animal (Sarnyai et al., 1995) and human studies (Gawin and Kleber, 1986), and

supports the hypothesis that cocaine withdrawal during adolescence significantly impairs

160

male rats risk-assessment. This is an extremely relevant result since impaired risk-

assessment may have important consequences in human adolescents.

Although elevated levels of novelty seeking (Adriani et al., 1998) and risk taking

(Macri et al., 2002), as well as reduced harm avoidance (Macri et al., 2002), are exhibited

by rodents during adolescence, our data seems to indicate that the adolescent high risk

typical behaviour was exacerbated during acute cocaine withdrawal. On the other hand,

such behavioural profile was not observed in female rats, which could be due to the fact

that, as in humans (Byrnes et al., 1999), male rats engage in more risk-taking behaviour

than females. In fact, marked sex differences were found before in mice, with males

showing significantly higher levels of novelty seeking than females (Palanza et al., 2001).

Another interesting observation is that in the cocaine exposed group, females

showed increased frequency of open arm entries, as well as an increase in the number of

closed arms entries and total arm entries. The increased frequency of closed and total

arm entries may reflect higher locomotor activity (Hogg, 1996). While increased frequency

of open arm entries is seen as an increase in anxiety levels in cocaine-treated females.

Therefore, an important sex difference was revealed by the EPM test, which may be

resultant from hormonal alterations induced by cocaine.

When looking at the possible effects of cocaine in the HPA axis it was seen that

plasma levels of corticosterone after the EPM, performed 2 days after withdrawal, were

not affected. Moreover, neuroendocrine basal data also showed no differences in basal

corticosterone plasma levels 1 day after cocaine withdrawal. These results may suggest

that the observed alterations on the EPM were not a consequence of an altered pattern of

corticosterone release.

A number of explanations can be presented for the absence of apparent

alterations of the HPA axis response during cocaine withdrawal observed in our study.

The rise in corticosterone after exposure to the novel environment provided by the EPM

apparatus could be so large that it obscures the drug treatment-induced alterations in the

increase of corticosterone (Schramm-Sapyta et al., 2006). On the other hand, age related

differences in activity of the HPA axis have been observed (Adriani and Laviola, 2000).

Adolescents have higher corticosterone levels at baseline than do adults, and such higher

baseline could contribute to a reduced percentage of increase compared to adults (Adriani

and Laviola, 2000; Schramm-Sapyta et al., 2006). Moreover, the HPA axis seems to be

somewhat hyporesponsive to external perturbations during adolescence. As shown in a

former study of response to forced novelty where acute amphetamine produce increase in

161

corticosterone release in adult mice, but only a trend toward slight elevation in adolescent

subjects (Adriani and Laviola, 2000).

Data from neurochemical analyses indicate that the DA and 5-HT metabolism were

altered in the prefrontal cortex of male rats chronically exposed to cocaine after the EPM,

as indicated by the increased DA and 5-HT turnover rates. In the amygdala there was also

an alteration on dopaminergic neurotransmission as indicated by the increased levels of

HVA levels in the same animals.

The dopaminergic system in the prefrontal cortex has been found to be involved in

the response to anxiogenic and stressfull situations in rats (Taylor et al., 1982). Decreases

in DA metabolism in the frontal cortex, but not striatum or nucleus accumbens, were

reported for up to 6 weeks following the discontinuation of chronic cocaine administration

in rats (Karoum et al., 1990), suggesting a persistent and specific down-regulation of DA

production in the frontal cortex (Goeders, 1997). Moreover, withdrawal from repeated

cocaine administration elicits some long-term neuroadaptations in the prefrontal cortex,

including reduced D2 receptor-mediated regulation of cortical excitability and reduced

responsivity of cortical cells to phasic increases in DA (Nogueira et al., 2006). The

prefrontal cortex is among the brain regions showing particularly marked ontogenetic

alterations during adolescence, with this region showing considerable synaptic culling and

a prominent loss of excitatory input (Bourgeois et al., 1994).

It is important to stress that, DA projections to the mesolimbic brain regions and

the prefrontal cortex are part of the circuitry critical for modulating risk taking, novelty

seeking, social behaviours (e.g., Le Moal and Simon, 1991) and motivational attaching to

reward (e.g., Robinson and Berridge, 2003). Because adolescents lack sufficient cortical

regulation, their behaviours tend to be more impulsive and guided by emotion impulses,

increasing risky-behaviour taking (Chambers et al., 2003). Moreover, it has been

demonstrated that prefrontocortical DA activation is necessary for coping with anxiogenic

challenges, allowing the animal to display adaptive behavioural responses (Espejo, 1997).

Therefore, the increased DA turnover rate in the prefrontal cortex we have observed, may

be involved in the impaired risk-assessment presented by cocaine exposed male rats

during the EPM.

The serotonergic system is associated with anxiety-related behaviour, with clinical

and animal studies showing increasing evidence that serotonergic neurotransmission in

the CNS modulates depression and anxiety-related behaviours (Eison, 1990; Millan et al.,

1997; van Megen et al., 1997). Results of microdialysis studies showed that aversive

conditions, such as the EPM and the conditioned fear-induced freezing behaviour, are

162

accompanied by increased 5-HT release in the prefrontal cortex (Yoshioka et al., 1995;

Inoue et al., 1996; Hashimoto et al., 1999) . Confirming this, the augmented 5-HT release

could be reduced by anxiolytic agents such as diazepam (Yoshioka et al., 1995). Rats

with lesions in the prefrontal cortex showed lower anxiety-related behaviour than their

sham-lesioned counterparts in social interaction and EPM (Gonzalez et al., 2000). Several

studies have been proposing a controlling role of 5-HT and the prefrontal cortex in

anxiety-related behaviour (Hashimoto et al., 1999; Gonzalez et al., 2000; Roberts and

Wallis, 2000), suggesting that the increase of 5-HT release in the prefrontal cortex under

aversive conditions does not mediate the “feeling” of anxiety but helps to cope with it.

Extracellular levels of 5-HT in the prefrontal cortex increases during conditioned fear-

induced freezing and this increase is followed by a resolution of the freezing (Hashimoto

et al., 1999). Therefore, the increased 5-HT turnover rate in the prefrontal cortex of

cocaine exposed male rats, which reflects a decreased availability of 5-HT, can also be

involved in the failure of these animals to cope with the challenge provided by the EPM,

suggesting, once again, a relation between cocaine-induced disruption of the prefrontal

cortex and the impaired risk-assessment.

The neurochemical data also showed an alteration in the dopaminergic system in

amygdala in chronically exposed males, as indicated by the increased levels of HVA

observed in this brain area. The amygdala has long been implicated in stimulus-reward

processes drug seeking behaviour. Exposure to a cocaine-related environment increased

the expression of c-fos in the amygdala (Neisewander et al., 2000), while amygdala

lesions disrupted acquisition of cocaine conditioned place preference (Fuchs et al., 2002).

More specifically, the amygdala dopaminergic system is involved in the cocaine self-

administration behaviour (McGregor and Roberts, 1993; Hurd et al., 1997; Hurd and

Ponten, 2000). Amygdala is also a critical structure in the neural system necessary for

implementing advantageous decisions. Amygdalar damage leads to decision-making

impairment, and this impairment is an indirect consequence of the role of the amygdala in

attaching affective attributes to stimuli (Bechara et al., 1999). It has been demonstrated

that monkeys with lesions of the amygdala have an increased tendency to approach

objects such as snakes (Zola-Morgan et al., 1991), as if the object of fear can no longer

evoke a state of fear (Bechara et al., 1999). In rats, numerous studies showed that

damaging different amygdala nuclei interferes with the processing of affective attributes of

reward stimuli (Hatfield et al., 1996). In the light of these data, the non-adaptive behaviour

observed in EPM in cocaine-treated males may be also a result of an inability to

experience sufficiently the emotional attributes of the situation provided by the EPM,

induce by an altered dopaminergic neurotransmission.

163

Adolescence is a period of increased impulsivity, which can be characterized by

decision making involving choices of high risks and low benefits (Chambers et al., 2003).

The perceived aversive consequences of risk-taking are usually reduced in adolescence

compared to young adulthood (Schramm-Sapyta et al., 2006). Recent evidence has linked

exposure to addictive drugs to an inability to use information about adverse consequences

or outcomes. For instance, addicts and drug-experienced animals fail to adapt their

behaviour to avoid adverse outcomes in gambling and reversal tasks or after changes in

the value of expected rewards (Grant et al., 2000; Bechara, 2001; Clark and Robbins,

2002).Our results do point to a cocaine-induced increase in the risk-taking behaviour in

adolescents male rats. This factor may contribute to the increased vulnerability of

adolescents to use drugs and to the maintenance of drug use, and therefore, these results

are not only biologically relevant, but also may contribute to the development of more

successful intervention strategies.

3.4.5. Effects of chronic cocaine treatment during adolescence on the depressive-like behaviour

Previous work in adult animals has shown that withdrawal from cocaine induces

depressive-like behaviour (Gawin and Kleber, 1986; Markou et al., 1992). Increased

passive behaviour responses in the FST such as immobility and decreased active

behaviours like swimming or struggling, are thought to be a clear indication of depressive-

like symptomatology (Detke et al., 1995; Lopez-Rubalcava and Lucki, 2000). However, in

our study these measures were not affected by cocaine treatment.

Following the criteria established to define immobility we were not able to observe

altered immobility behaviour in none of the studied group of animals. Former works have

shown that there was an age-dependent difference in the amount of time spent in

immobility in a forced swim test (Bilitzke and Church, 1992; Molina et al., 1994), with

juveniles showing very little evidence of immobility in contrast to adults (Hansen-Trench

and Barron, 2005). However, at least one previous study reported immobility response in

adolescent rats (Cannizzaro et al., 2007). The absence of immobility behaviour observed

could be due to methodological differences in experimental procedures, such as

apparatus dimensions, water temperature, and/or criteria for defining immobility. However,

it should be noted that there were no treatment effects in the time spent struggling in both

males and females, which allow us to propose that the behavioural strategy adopted to

cope with the stressful condition provide by the FST was not affected by the cocaine

treatment and withdrawal. The absence of significant differences between control and

164

cocaine exposed rats, may be related with the reduced behavioural and physiological

response to stressful situations in adolescent rodents (Laviola et al., 2002; Macri et al.,

2002). On the other hand, to perform hormonal measurements our rats had to be housed

in isolation and despite our efforts to minimize depressive effects through improved

handling, the isolation could interfere with the outcomes in the FST, hiding possible effects

of cocaine treatment regarding a depression-like behaviour development.

Beside the evaluation of the behavioural response to FST, alterations in

physiological responses to the stressful situation provide by the FST during withdrawal

from chronic cocaine administration were also analysed. Like in the EPM, corticosterone

levels after the FST were not significantly increased in treated animals, which supports the

already discussed possibilities: a large rise in corticosterone after exposure to the novel

environment provided by the test may obscures a possible drug treatment-induced rise in

corticosterone; the elevated basal levels of corticosterone present in adolescents or the

age-related discontinuities in the response of the HPA axis to both stress and

psychostimulants could also be reasons for the absence of alterations of the HPA axis

response to FST.

The neurochemical data indicated that the DA turnover rate in SN-VTA was higher

in cocaine-treated male rats as compared with control males. It is known that response to

stress implicates alterations in dopaminergic activity (Watanabe, 1984; Kalivas and

Abhold, 1987; Castro and Zigmond, 2001), thus an inadequate dopaminergic response to

stress may lead to a non adaptive response to a stressful stimulus. However, despite the

effect of cocaine treatment in the dopaminergic neurotransmission in the SN-VTA, no

effects of cocaine treatment were observed in the strategy adopted to cope with the

stressful situation provided by the FST.

Regarding the serotonergic system, we have determined, in the amygdala, within

the control group an increase in the 5-HT turnover rate males. The increase in

serotonergic activity has been reported in response to other stressors (Dunn, 1988; Inoue

et al., 1994). In fact, such effect has already been demonstrated in the amygdala of adult

male rats as a result of FST exposure (Connor et al., 1997). Nevertheless, data presented

in here reveals a sex effect in the serotonergic activity in control animals that was not

observed in cocaine exposed rats, which may be a consequence of cocaine treatment and

its withdrawal.

Together, these results seem to show that cocaine treatment during the

adolescence did affect the neurotransmitter systems response to the FST.

165

In conclusion, although withdrawal from chronic cocaine administration during

adolescence seems not to have a significant effect on stress-induced behavioural

alterations, neurochemical data indicate that the response to the stressful situation

provided by the FST was affected. These results are substantially different from the data

obtained in studies carried out during the adulthood, which do showed stronger influence

of cocaine in stress response (Collins and Izenwasser, 2002; O'Dell et al., 2007). These

provides further evidence supporting previous findings suggesting that adolescents

respond differently from older animals to both stress and psychostimulants (Infurna and

Spear, 1979; Adriani and Laviola, 2000; Stansfield and Kirstein, 2005). This age related

effect could be one of the reasons for the elevated prevalence of drug use and

dependence during adolescence (Schramm-Sapyta et al., 2006). Indeed, age-related

discontinuities in response to psychological stress have been indicated as a

psychobiological risk for increased vulnerability to drug abuse (Laviola et al., 1995; Adriani

et al., 1998).

3.5. Conclusion

Overall, our data lead us to conclude that the chronic exposure to cocaine

throughout adolescence produced a sex-dependent hormonal, neurochemical and

behavioural alterations.

Acute cocaine binge administration after 15 days of treatment increased the HPA

activity in male rats, as indicated by the elevated ACTH levels observed 30 minutes after

cocaine exposure. However, the HPA axis activity, evaluated 1 day after the last

administration of cocaine, did not showed to be affected in neither males nor females.

Although HPA axis activation during cocaine withdrawal has been previously

demonstrated with a variety of paradigms, our data support the idea that age-related

discontinuity in the HPA axis function may be underlying a hypoactivation of the HPA axis

during the adolescence period.

Regarding the HPG axis activity, chronic cocaine exposure throughout

adolescence also led to short-term disruption also in male rats, as indicated by the

decreased testosterone levels 30 minutes after the last administration of cocaine.

Although 1 day after withdrawal this alteration was no longer observed our long-term data

(see chapter 4) reveals important changes in the HPG axis function at later stages. In

adolescent female rats the chronic binge cocaine administration seems not to affect the

HPG axis as observed through unaltered estradiol or progesterone levels.

166

Neurochemical data revealed the development of tolerance in the dopaminergic

and serotonergic responses to the acute effects of cocaine administration.

Data obtained 1 day after the last administration, revealed an increased

dopaminergic activity, which led us to suggest that the expected decrease in

dopaminergic activity after chronic cocaine, could have been masked by the cocaine

cues-induced increases in dopaminergic function, since animals were sacrificed in the

same room where cocaine was administered. This may indicate a high susceptibility of

adolescent rats to environmental cues, with females showing a greater involvement of

amygdala in this process, while males showed a greater involvement of the nigrostriatal

pathway.

In the EPM it was observed an impaired risk-assessment in male rats. The

disrupted risk-assessment may be related with the high rates of prevalence of drug uses

during adolescence, and with increased vulnerability to relapse to drug use after cocaine

exposure throughout this developmental period. Along with impaired risk-assessment it

was observed a disrupted dopaminergic and serotonergic transmission in prefrontal cortex

and dopaminergic neurotransmission in amygdala. These neurochemical alterations may

be involved in the failure of cocaine-treated male rats to exhibit an adaptive behaviour

during the EPM.

In agreement, the 5-HT and DA response to the FST, in male rats, seems to

support the inability of animals to cope with stress.

In conclusion, the adaptive processes developed in dopaminergic and serotonergic

responses, the elevated susceptibility to the cocaine-cues, the observed impairment in

risk-assessment and the absence of depressive behaviour after cocaine treatment

throughout adolescence, are all most likely important factors contributing to the

adolescent high vulnerability to drug abuse.

These data, have increased relevance for the social-understanding of the

relationship between adolescents and drug use, and may be crucial for the design of new

strategies of drug abuse prevention in the adolescent.

167

3.6. References

Adell A, Carceller A, Artigas F (1991) Regional distribution of extracellular 5-hydroxytryptamine and 5-hydroxyindoleacetic acid in the brain of freely moving rats. J Neurochem 56:709-712.

Adriani W, Laviola G (2000) A unique hormonal and behavioral hyporesponsivity to both forced novelty and d-amphetamine in periadolescent mice. Neuropharmacology 39:334-346.

Adriani W, Chiarotti F, Laviola G (1998) Elevated novelty seeking and peculiar d-amphetamine sensitization in periadolescent mice compared with adult mice. Behavioral Neuroscience 112:1152-1166.

Aloisi AM, Steenbergen HL, van de Poll NE, Farabollini F (1994) Sex-dependent effects of restraint on nociception and pituitary-adrenal hormones in the rat. Physiol Behav 55:789-793.

Andersen SL, Gazzara RA (1993) The ontogeny of apomorphine-induced alterations of neostriatal dopamine release: effects on spontaneous release. J Neurochem 61:2247-2255.

Andersen SL, Thompson AP, Krenzel E, Teicher MH (2002) Pubertal changes in gonadal hormones do not underlie adolescent dopamine receptor overproduction. Psychoneuroendocrinology 27:683-691.

Anderson SM, Saviolakis GA, Bauman RA, Chu KY, Ghosh S, Kant GJ (1996) Effects of chronic stress on food acquisition, plasma hormones, and the estrous cycle of female rats. Physiol Behav 60:325-329.

Antkiewicz-Michaluk L, Filip M, Michaluk J, Romanska I, Patsenka A, Papla I, Przegalinski E, Vetulani J (2006) Conditioned rewarding stimulus associated with cocaine self-administration reverses the depression of catecholamine brain systems following cocaine withdrawal in rats. Int J Neuropsychopharmacol 9:37-50.

Arpin-Bott MP, Dietrich JB, Dirrig-Grosch S, Aunis D, Zwiller J (2006) Induction by cocaine of the serotonergic 5-HT3 receptor in rat cerebellum. Ann N Y Acad Sci 1074:382-389.

Atkinson HC, Waddell BJ (1997) Circadian variation in basal plasma corticosterone and adrenocorticotropin in the rat: sexual dimorphism and changes across the estrous cycle. Endocrinology 138:3842-3848.

Azmitia EC, Mcnaughton N, Tsaltas L, Fillenz M, Gray JA (1984) Interactions between Hippocampal Serotonin and the Pituitary-Adrenal Axis in the Septal Driving of Hippocampal Theta-Rhythm. Neuroendocrinology 39:471-475.

Badanich KA, Adler KJ, Kirstein CL (2006) Adolescents differ from adults in cocaine conditioned place preference and cocaine-induced dopamine in the nucleus accumbens septi. Eur J Pharmacol 550:95-106.

Badiani A, Browman KE, Robinson TE (1995) Influence of novel versus home environments on sensitization to the psychomotor stimulant effects of cocaine and amphetamine. Brain Res 674:291-298.

Baker S, Bielajew C (2007) Influence of housing on the consequences of chronic mild stress in female rats. Stress 10:283-293.

Balopole DC, Hansult CD, Dorph D (1979) Effect of cocaine on food intake in rats. Psychopharmacology (Berl) 64:121-122.

Banks ML, Czoty PW, Gage HD, Bounds MC, Garg PK, Garg S, Nader MA (2008) Effects of cocaine and MDMA self-administration on serotonin transporter availability in monkeys. Neuropsychopharmacology 33:219-225.

Barron S, White A, Swartzwelder HS, Bell RL, Rodd ZA, Slawecki CJ, Ehlers CL, Levin ED, Rezvani AH, Spear LP (2005) Adolescent vulnerabilities to chronic alcohol or nicotine exposure: findings from rodent models. Alcohol Clin Exp Res 29:1720-1725.

Barroso-Moguel R, Mendez-Armenta M, Villeda-Hernandez J (1994) Testicular lesions by chronic administration of cocaine in rats. J Appl Toxicol 14:37-41.

168

Barroso-Moguel R, Mendez-Armenta M, Villeda-Hernandez J, Nava-Ruiz C, Santamaria A (2002) Brain lesions induced by chronic cocaine administration to rats. Prog Neuropsychopharmacol Biol Psychiatry 26:59-63.

Basso AM, Vale W, Koob GF, Spina M, Rivier J (1999) Corticotropin-releasing factor antagonist attenuates the "anxiogenic-like" effect in the defensive burying paradigm but not in the elevated plus-maze following chronic cocaine in rats. Psychopharmacology (Berl) 145:21-30.

Baumann MH, Becketts KM, Rothman RB (1995) Evidence for alterations in presynaptic serotonergic function during withdrawal from chronic cocaine in rats. Eur J Pharmacol 282:87-93.

Baumann MH, Milchanowski AB, Rothman RB (2004) Evidence for alterations in alpha2-adrenergic receptor sensitivity in rats exposed to repeated cocaine administration. Neuroscience 125:683-690.

Baumann MH, Raley TJ, Partilla JS, Rothman RB (1993) Biosynthesis of dopamine and serotonin in the rat brain after repeated cocaine injections: a microdissection mapping study. Synapse 14:40-50.

Bechara A (2001) Neurobiology of decision-making: risk and reward. Semin Clin Neuropsychiatry 6:205-216.

Bechara A, Damasio H, Damasio AR, Lee GP (1999) Different contributions of the human amygdala and ventromedial prefrontal cortex to decision-making. J Neurosci 19:5473-5481.

Beitner-Johnson D, Nestler EJ (1991) Morphine and cocaine exert common chronic actions on tyrosine hydroxylase in dopaminergic brain reward regions. J Neurochem 57:344-347.

Benloucif S, Galloway MP (1991) Facilitation of dopamine release in vivo by serotonin agonists: studies with microdialysis. Eur J Pharmacol 200:1-8.

Berul CI, Harclerode JE (1989) Effects of cocaine hydrochloride on the male reproductive system. Life Sci 45:91-95.

Bilitzke PJ, Church MW (1992) Prenatal cocaine and alcohol exposures affect rat behavior in a stress test (the Porsolt swim test). Neurotoxicol Teratol 14:359-364.

Bishop GA, Ho RH (1985) The distribution and origin of serotonin immunoreactivity in the rat cerebellum. Brain Res 331:195-207.

Bissiere S, Humeau Y, Luthi A (2003) Dopamine gates LTP induction in lateral amygdala by suppressing feedforward inhibition. Nat Neurosci 6:587-592.

Bjork JM, Knutson B, Fong GW, Caggiano DM, Bennett SM, Hommer DW (2004) Incentive-elicited brain activation in adolescents: similarities and differences from young adults. J Neurosci 24:1793-1802.

Blandina P, Goldfarb J, Craddock-Royal B, Green JP (1989) Release of endogenous dopamine by stimulation of 5-hydroxytryptamine3 receptors in rat striatum. J Pharmacol Exp Ther 251:803-809.

Bolanos C, Glatt S, Jackson D (1998) Subsensitivity to dopaminergic drugs in periadolescent rats: a behavioral and neurochemical analysis. Brain Res Dev Brain Res 111:25-33.

Borowsky B, Kuhn CM (1991) Monoamine mediation of cocaine-induced hypothalamo-pituitary-adrenal activation. J Pharmacol Exp Ther 256:204-210.

Bourgeois JP, Goldman-Rakic PS, Rakic P (1994) Synaptogenesis in the prefrontal cortex of rhesus monkeys. Cereb Cortex 4:78-96.

Bradberry CW, Nobiletti JB, Elsworth JD, Murphy B, Jatlow P, Roth RH (1993) Cocaine and cocaethylene: microdialysis comparison of brain drug levels and effects on dopamine and serotonin. J Neurochem 60:1429-1435.

169

Brenhouse HC, Andersen SL (2008) Delayed extinction and stronger reinstatement of cocaine conditioned place preference in adolescent rats, compared to adults. Behav Neurosci 122:460-465.

Brinley-Reed M, McDonald AJ (1999) Evidence that dopaminergic axons provide a dense innervation of specific neuronal subpopulations in the rat basolateral amygdala. Brain Res 850:127-135.

Brock JW, Ng JP, Justice JB, Jr. (1990) Effect of chronic cocaine on dopamine synthesis in the nucleus accumbens as determined by microdialysis perfusion with NSD-1015. Neurosci Lett 117:234-239.

Brodie MS, Bunney EB (1996) Serotonin potentiates dopamine inhibition of ventral tegmental area neurons in vitro. J Neurophysiol 76:2077-2082.

Brown JM, Riddle EL, Sandoval V, Weston RK, Hanson JE, Crosby MJ, Ugarte YV, Gibb JW, Hanson GR, Fleckenstein AE (2002) A single methamphetamine administration rapidly decreases vesicular dopamine uptake. J Pharmacol Exp Ther 302:497-501.

Brudzynski SM, Gibson CJ (1997) Release of dopamine in the nucleus accumbens caused by stimulation of the subiculum in freely moving rats. Brain Res Bull 42:303-308.

Buckingham JC, Hodges JR (1979) Hypothalamic receptors influencing the secretion of corticotrophin releasing hormone in the rat. J Physiol 290:421-431.

Budziszewska B, Jaworska-Feil L, Lason W (1996) The effect of repeated amphetamine and cocaine administration on adrenal, gonadal and thyroid hormone levels in the rat plasma. Exp Clin Endocrinol Diabetes 104:334-338.

Bull DR, Bakhtiar R, Sheehan MJ (1991) Characterization of dopamine autoreceptors in the amygdala: a fast cyclic voltammetric study in vitro. Neurosci Lett 134:41-44.

Byrnes JP, Miller DC, Schafer WD (1999) Gender differences in risk taking: A meta-analysis. Psychological Bulletin 125:367-383.

Cameron DL, Wessendorf MW, Williams JT (1997) A subset of ventral tegmental area neurons is inhibited by dopamine, 5-hydroxytryptamine and opioids. Neuroscience 77:155-166.

Cannizzaro C, Plescia F, Gagliano M, Cannizzaro G, Mantia G, La Barbera M, Provenzano G, Cannizzaro E (2007) Perinatal exposure to 5-metoxytryptamine, behavioural-stress reactivity and functional response of 5-HT1A receptors in the adolescent rat. Behav Brain Res 186 (1):98-106.

Castro SL, Zigmond MJ (2001) Stress-induced increase in extracellular dopamine in striatum: role of glutamatergic action via N-methyl-D-aspartate receptors in substantia nigra. Brain Res 901:47-54.

Chambers RA, Taylor JR, Potenza MN (2003) Developmental neurocircuitry of motivation in adolescence: A critical period of addiction vulnerability. Am J Psychiatry 160:1041-1052.

Chefer VI, Moron JA, Hope B, Rea W, Shippenberg TS (2000) Kappa-opioid receptor activation prevents alterations in mesocortical dopamine neurotransmission that occur during abstinence from cocaine. Neuroscience 101:619-627.

Chen K, Kandel DB (1995) The natural history of drug use from adolescence to the mid-thirties in a general population sample. Am J Public Health 85:41-47.

Chen K, Kandel DB, Davies M (1997) Relationships between frequency and quantity of marijuana use and last year proxy dependence among adolescents and adults in the United States. Drug Alcohol Depend 46:53-67.

Childress AR, Mozley PD, McElgin W, Fitzgerald J, Reivich M, O'Brien CP (1999) Limbic activation during cue-induced cocaine craving. Am J Psychiatry 156:11-18.

Chisari A, Carino M, Perone M, Gaillard RC, Spinedi E (1995) Sex and strain variability in the rat hypothalamo-pituitary-adrenal (HPA) axis function. J Endocrinol Invest 18:25-33.

170

Ciccocioppo R, Sanna PP, Weiss F (2001) Cocaine-predictive stimulus induces drug-seeking behavior and neural activation in limbic brain regions after multiple months of abstinence: reversal by D(1) antagonists. Proc Natl Acad Sci USA 98:1976-1981.

Clark DB, Kirisci L, Tarter RE (1998) Adolescent versus adult onset and the development of substance use disorders in males. Drug Alcohol Depend 49:115-121.

Clark L, Robbins T (2002) Decision-making deficits in drug addiction. Trends Cogn Sci 6:361.

Claye LH, Akunne HC, Davis MD, DeMattos S, Soliman KF (1995) Behavioral and neurochemical changes in the dopaminergic system after repeated cocaine administration. Mol Neurobiol 11:55-66.

Cocores JA, Dackis CA, Gold MS (1986) Sexual dysfunction secondary to cocaine abuse in two patients. J Clin Psychiatry 47:384-385.

Collins SL, Izenwasser S (2002) Cocaine differentially alters behavior and neurochemistry in periadolescent versus adult rats. Brain Res Dev Brain Res 138:27-34.

Connor TJ, Kelly JP, Leonard BE (1997) Forced swim test-induced neurochemical endocrine, and immune changes in the rat. Pharmacol Biochem Behav 58:961-967.

Cooper SJ, van der Hoek GA (1993) Cocaine: a microstructural analysis of its effects on feeding and associated behaviour in the rat. Brain Res 608:45-51.

Cowan RL, Sesack SR, Van Bockstaele EJ, Branchereau P, Chain J, Pickel VM (1994) Analysis of synaptic inputs and targets of physiologically characterized neurons in rat frontal cortex: combined in vivo intracellular recording and immunolabeling. Synapse 17:101-114.

Cunningham K, Callahan P (1991) Monoamine reuptake inhibitors enhance the discriminative state induced by cocaine in the rat. Psychopharmacology (Berl) 104:177-180.

Cunningham KA, Paris JM, Goeders NE (1992) Chronic cocaine enhances serotonin autoregulation and serotonin uptake binding. Synapse 11:112-123.

Cunningham KA, Bradberry CW, Chang AS, Reith ME (1996) The role of serotonin in the actions of psychostimulants: molecular and pharmacological analyses. Behav Brain Res 73:93-102.

Cunningham MG, Bhattacharyya S, Benes FM (2002) Amygdalo-cortical sprouting continues into early adulthood: implications for the development of normal and abnormal function during adolescence. J Comp Neurol 453:116-130.

Dackis CA, Gold MS (1985) New concepts in cocaine addiction: the dopamine depletion hypothesis. Neurosci Biobehav Rev 9:469-477.

Daunais JB, McGinty JF (1995) Cocaine binges differentially alter striatal preprodynorphin and zif/268 mRNAs. Brain Res Mol Brain Res 29:201-210.

de Jong IE, Oitzl MS, de Kloet ER (2007) Adrenalectomy prevents behavioural sensitisation of mice to cocaine in a genotype-dependent manner. Behav Brain Res 177:329-339.

De Martino B, Kumaran D, Seymour B, Dolan RJ (2006) Frames, biases, and rational decision-making in the human brain. Science 313:684-687.

Detke MJ, Rickels M, Lucki I (1995) Active behaviors in the rat forced swimming test differentially produced by serotonergic and noradrenergic antidepressants. Psychopharmacology (Berl) 121:66-72.

Di Chiara G, Bassareo V, Fenu S, De Luca MA, Spina L, Cadoni C, Acquas E, Carboni E, Valentini V, Lecca D (2004) Dopamine and drug addiction: the nucleus accumbens shell connection. Neuropharmacology 47 Suppl 1:227-241.

Di Ciano P, Everitt BJ (2004) Direct interactions between the basolateral amygdala and nucleus accumbens core underlie cocaine-seeking behavior by rats. J Neurosci 24:7167-7173.

Drouin J, Labrie F (1976) Selective effect of androgens on LH and FSH release in anterior pituitary cells in culture. Endocrinology 98:1528-1534.

Dunn AJ (1988) Stress-related changes in cerebral catecholamine and indoleamine metabolism: lack of effect of adrenalectomy and corticosterone. J Neurochem 51:406-412.

171

Duvauchelle CL, Ikegami A, Castaneda E (2000) Conditioned increases in behavioral activity and accumbens dopamine levels produced by intravenous cocaine. Behav Neurosci 114:1156-1166.

Dwoskin LP, Peris J, Yasuda RP, Philpott K, Zahniser NR (1988) Repeated cocaine administration results in supersensitivity of striatal D-2 dopamine autoreceptors to pergolide. Life Sci 42:255-262.

Eison MS (1990) Serotonin: a common neurobiologic substrate in anxiety and depression. J Clin Psychopharmacol 10:26S-30S.

Espejo EF (1997) Selective dopamine depletion within the medial prefrontal cortex induces anxiogenic-like effects in rats placed on the elevated plus maze. Brain Research 762:281-284.

Estelles, Aguilar MA, Miñarro J, Lluch J, Rodríguez-Arias M (2007) Cocaine exposure during adolescence affects anxiety in adult mice. Brain Res Bull 71:393-403.

Estroff TW, Schwartz RH, Hoffmann NG (1989) Adolescent cocaine abuse- addictive potential, behavioral and psychiatric effects. Clinical Pediatrics 28:550-555.

Feder HH, Ruf KB (1969) Stimulation of progesterone release and estrous behavior by ACTH in ovariectomized rodents. Endocrinology 84:171-174.

Ferre S, Cortes R, Artigas F (1994) Dopaminergic regulation of the serotonergic raphe-striatal pathway: microdialysis studies in freely moving rats. J Neurosci 14:4839-4846.

Freund TF, Antal M (1988) GABA-containing neurons in the septum control inhibitory interneurons in the hippocampus. Nature 336:170-173.

Freund TF, Gulyas AI, Acsady L, Gorcs T, Toth K (1990) Serotonergic control of the hippocampus via local inhibitory interneurons. Proc Natl Acad Sci U S A 87:8501-8505.

Frey U, Schroeder H, Matthies H (1990) Dopaminergic antagonists prevent long-term maintenance of posttetanic LTP in the CA1 region of rat hippocampal slices. Brain Res 522:69-75.

Fuchs RA, Weber SM, Rice HJ, Neisewander JL (2002) Effects of excitotoxic lesions of the basolateral amygdala on cocaine-seeking behavior and cocaine conditioned place preference in rats. Brain Res 929:15-25.

Gabbott PL, Warner TA, Busby SJ (2006) Amygdala input monosynaptically innervates parvalbumin immunoreactive local circuit neurons in rat medial prefrontal cortex. Neuroscience 139:1039-1048.

Gasbarri A, Packard MG, Campana E, Pacitti C (1994) Anterograde and retrograde tracing of projections from the ventral tegmental area to the hippocampal formation in the rat. Brain Res Bull 33:445-452.

Gawin FH (1991) Cocaine addiction: psychology and neurophysiology. Science 251:1580-1586.

Gawin FH, Kleber HD (1986) Abstinence symptomatology and psychiatric diagnosis in cocaine abusers. Clinical observations. Arch Gen Psychiatry 43:107-113.

Gawin FH, Kleber HD, Byck R, Rounsaville BJ, Kosten TR, Jatlow PI, Morgan C (1989) Desipramine facilitation of initial cocaine abstinence. Arch Gen Psychiatry 46:117-121.

George VK, Li H, Teloken C, Grignon DJ, Lawrence WD, Dhabuwala CB (1996) Effects of long-term cocaine exposure on spermatogenesis and fertility in peripubertal male rats. J Urol 155:327-331.

Ghitza UE, Rothman RB, Gorelick DA, Henningfield JE, Baumann MH (2007) Serotonergic responsiveness in human cocaine users. Drug Alcohol Depend 86:207-213.

Giedd JN, Blumenthal J, Jeffries NO, Castellanos FX, Liu H, Zijdenbos A, Paus T, Evans AC, Rapoport JL (1999) Brain development during childhood and adolescence: a longitudinal MRI study. Nat Neurosci 2:861-863.

Giorgetti M, Zhdanova IV (2000) Chronic cocaine treatment induces dysregulation in the circadian pattern of rats' feeding behavior. Brain Res 877:170-175.

172

Glaser PE, Surgener SP, Grondin R, Gash CR, Palmer M, Castellanos FX, Gerhardt GA (2006) Cerebellar neurotransmission in attention-deficit/hyperactivity disorder: does dopamine neurotransmission occur in the cerebellar vermis? J Neurosci Methods 151:62-67.

Goeders N (1997) A neuroendocrine role in cocaine reinforcement. Psychoneuroendocrinology 22:237-259.

Goeders NE, McNulty MA, Guerin GF (1993) Effects of alprazolam on intravenous cocaine self-administration in rats. Pharmacology Biochemistry and Behavior 44:471-474.

Gomez F, Dallman MF (2001) Manipulation of androgens causes different energetic responses to cold in 60- and 40-day-old male rats. Am J Physiol Regul Integr Comp Physiol 280:R262-273.

Gomez F, Houshyar H, Dallman MF (2002) Marked regulatory shifts in gonadal, adrenal, and metabolic system responses to repeated restraint stress occur within a 3-week period in pubertal male rats. Endocrinology 143:2852-2862.

Gonzalez LE, Rujano M, Tucci S, Paredes D, Silva E, Alba G, Hernandez L (2000) Medial prefrontal transection enhances social interaction. I: behavioral studies. Brain Res 887:7-15.

Gore AC, Attardi B, DeFranco DB (2006) Glucocorticoid repression of the reproductive axis: effects on GnRH and gonadotropin subunit mRNA levels. Mol Cell Endocrinol 256:40-48.

Gottschalk PC, Kosten TR (2002) Cerebral perfusion defects in combined cocaine and alcohol dependence. Drug Alcohol Depend 68:95-104.

Gottwald B, Wilde B, Mihajlovic Z, Mehdorn HM (2004) Evidence for distinct cognitive deficits after focal cerebellar lesions. J Neurol Neurosurg Psychiatry 75:1524-1531.

Goussakov I, Chartoff EH, Tsvetkov E, Gerety LP, Meloni EG, Carlezon WA, Jr., Bolshakov VY (2006) LTP in the lateral amygdala during cocaine withdrawal. Eur J Neurosci 23:239-250.

Grant S, Contoreggi C, London ED (2000) Drug abusers show impaired performance in a laboratory test of decision making. Neuropsychologia 38:1180-1187.

Grant S, London ED, Newlin DB, Villemagne VL, Liu X, Contoreggi C, Phillips RL, Kimes AS, Margolin A (1996) Activation of memory circuits during cue-elicited cocaine craving. Proc Natl Acad Sci USA 93:12040-12045.

Grobin AC, Deutch AY (1998) Dopaminergic regulation of extracellular gamma-aminobutyric acid levels in the prefrontal cortex of the rat. J Pharmacol Exp Ther 285:350-357.

Gulyas AI, Acsady L, Freund TF (1999) Structural basis of the cholinergic and serotonergic modulation of GABAergic neurons in the hippocampus. Neurochem Int 34:359-372.

Haj-Dahmane S (2001) D2-like dopamine receptor activation excites rat dorsal raphe 5-HT neurons in vitro. Eur J Neurosci 14:125-134.

Hallbus M, Magnusson T, Magnusson O (1997) Influence of 5-HT1B/1D receptors on dopamine release in the guinea pig nucleus accumbens: a microdialysis study. Neurosci Lett 225:57-60.

Handa RJ, Burgess LH, Kerr JE, O'Keefe JA (1994) Gonadal steroid hormone receptors and sex differences in the hypothalamo-pituitary-adrenal axis. Horm Behav 28:464-476.

Hanley NR, Van de Kar LD (2003) Serotonin and the neuroendocrine regulation of the hypothalamic--pituitary-adrenal axis in health and disease. Vitam Horm 66:189-255.

Hansen-Trench LS, Barron S (2005) Effects of neonatal alcohol and/or cocaine exposure on stress in juvenile and adult female rats. Neurotoxicol Teratol 27:55-63.

Harris GC, Aston-Jones G (1993) Beta-adrenergic antagonists attenuate withdrawal anxiety in cocaine- and morphine-dependent rats. Psychopharmacology (Berl) 113:131-136.

Harrison PA, Hoffmann NG, Edwall GE (1989) Differential drug use patterns among sexually abused adolescent girls in treatment for chemical dependency. Int J Addict 24:499-514.

173

Hashimoto S, Inoue T, Koyama T (1999) Effects of conditioned fear stress on serotonin neurotransmission and freezing behavior in rats. Eur J Pharmacol 378:23-30.

Hatfield T, Han JS, Conley M, Gallagher M, Holland P (1996) Neurotoxic lesions of basolateral, but not central, amygdala interfere with Pavlovian second-order conditioning and reinforcer devaluation effects. J Neurosci 16:5256-5265.

Haycock JW, Becker L, Ang L, Furukawa Y, Hornykiewicz O, Kish SJ (2003) Marked disparity between age-related changes in dopamine and other presynaptic dopaminergic markers in human striatum. J Neurochem 87:574-585.

Hester R, Garavan H (2004) Executive dysfunction in cocaine addiction: evidence for discordant frontal, cingulate, and cerebellar activity. J Neurosci 24:11017-11022.

Hogg S (1996) A review of the validity and variability of the elevated plus-maze as an animal model of anxiety. Pharmacol Biochem Behav 54:21-30.

Horne MK, Lee J, Chen F, Lanning K, Tomas D, Lawrence AJ (2008) Long-term administration of cocaine or serotonin reuptake inhibitors results in anatomical and neurochemical changes in noradrenergic, dopaminergic, and serotonin pathways. Journal of Neurochemistry 106:1731-1744.

Hurd YL, Ungerstedt U (1989) Cocaine: an in vivo microdialysis evaluation of its acute action on dopamine transmission in rat striatum. Synapse 3:48-54.

Hurd YL, Ponten M (2000) Cocaine self-administration behavior can be reduced or potentiated by the addition of specific dopamine concentrations in the nucleus accumbens and amygdala using in vivo microdialysis. Behav Brain Res 116:177-186.

Hurd YL, McGregor A, Ponten M (1997) In vivo amygdala dopamine levels modulate cocaine self-administration behaviour in the rat: D1 dopamine receptor involvement. Eur J Neurosci 9:2541-2548.

Imperato A, Mele A, Scrocco MG, Puglisi-Allegra S (1992) Chronic cocaine alters limbic extracellular dopamine. Neurochemical basis for addiction. Eur J Pharmacol 212:299-300.

Infurna RN, Spear LP (1979) Developmental changes in amphetamine-induced taste aversions. Pharmacol Biochem Behav 11:31-35.

Inoue T, Tsuchiya K, Koyama T (1994) Regional changes in dopamine and serotonin activation with various intensity of physical and psychological stress in the rat brain. Pharmacol Biochem Behav 49:911-920.

Inoue T, Tsuchiya K, Koyama T (1996) Serotonergic activation reduces defensive freezing in the conditioned fear paradigm. Pharmacol Biochem Behav 53:825-831.

Iyer RN, Bradberry CW (1996) Serotonin-mediated increase in prefrontal cortex dopamine release: pharmacological characterization. J Pharmacol Exp Ther 277:40-47.

Jacobs BL, Azmitia EC (1992) Structure and Function of the Brain-Serotonin System. Physiological Reviews 72:165-229.

Jacobsen LK, Staley JK, Malison RT, Zoghbi SS, Seibyl JP, Kosten TR, Innis RB (2000) Elevated central serotonin transporter binding availability in acutely abstinent cocaine-dependent patients. American Journal of Psychiatry 157:1134-1140.

Johnson RG, Fiorella D, Rabin RA (1993) Effects of chronic cocaine administration on the serotonergic system in the rat brain. Pharmacol Biochem Behav 46:289-293.

Jones S, Bonci A (2005) Synaptic plasticity and drug addiction. Curr Opin Pharmacol 5:20-25.

Kalivas PW, Abhold R (1987) Enkephalin release into the ventral tegmental area in response to stress: modulation of mesocorticolimbic dopamine. Brain Res 414:339-348.

Kalivas PW, Duffy P (1990) Effect of acute and daily cocaine treatment on extracellular dopamine in the nucleus accumbens. Synapse 5:48-58.

Kalra SP, Kalra PS (1983) Neural regulation of luteinizing hormone secretion in the rat. Endocr Rev 4:311-351.

174

Karoum F, Suddath RL, Wyatt RJ (1990) Chronic cocaine and rat brain catecholamines: long-term reduction in hypothalamic and frontal cortex dopamine metabolism. Eur J Pharmacol 186:1-8.

Kelland MD, Freeman AS, Chiodo LA (1990) Serotonergic afferent regulation of the basic physiology and pharmacological responsiveness of nigrostriatal dopamine neurons. J Pharmacol Exp Ther 253:803-811.

Kerstetter KA, Kantak KM (2007) Differential effects of self-administered cocaine in adolescent and adult rats on stimulus-reward learning. Psychopharmacology (Berl) 194:403-411.

King T, Schenken R, Kang I, Javors M, Riehl R (1990) Cocaine disrupts estrous cyclicity and alters the reproductive neuroendocrine axis in the rat. Neuroendocrinology 51:15-22.

King TS, Canez MS, Gaskill S, Javors MA, Schenken RS (1993) Chronic cocaine disruption of estrous cyclicity in the rat: dose-dependent effects. J Pharmacol Exp Ther 264:29-34.

Kitahama K, Nagatsu I, Geffard M, Maeda T (2000) Distribution of dopamine-immunoreactive fibers in the rat brainstem. J Chem Neuroanat 18:1-9.

Kiyatkin EA, Stein EA (1996) Conditioned changes in nucleus accumbens dopamine signal established by intravenous cocaine in rats. Neurosci Lett 211:73-76.

Koeltzow TE, White FJ (2003) Behavioral depression during cocaine withdrawal is associated with decreased spontaneous activity of ventral tegmental area dopamine neurons. Behav Neurosci 117:860-865.

Koob G, Sanna P, Bloom F (1998) Neuroscience of addiction. Neuron 21:467-476.

Krettek JE, Price JL (1977) Projections from the amygdaloid complex to the cerebral cortex and thalamus in the rat and cat. J Comp Neurol 172:687-722.

Kuhn C, Francis R (1997) Gender difference in cocaine-induced HPA axis activation. Neuropsychopharmacology 16:399-407.

Laviola G, Pascucci T, Pieretti S (2001) Striatal dopamine sensitization to D-amphetamine in periadolescent but not in adult rats. Pharmacol Biochem Behav 68:115-124.

Laviola G, Adriani W, Terranova ML, Gerra G (1999) Psychobiological risk factors for vulnerability to psychostimulants in human adolescents and animal models. Neuroscience and Biobehavioral Reviews 23:993-1010.

Laviola G, Adriani W, Morley-Fletcher S, Terranova ML (2002) Peculiar response of adolescent mice to acute and chronic stress and to amphetamine: evidence of sex differences. Behav Brain Res 130:117-125.

Laviola G, Wood RD, Kuhn C, Francis R, Spear LP (1995) Cocaine sensitization in periadolescent and adult rats. J Pharmacol Exp Ther 275:345-357.

Le Moal M, Simon H (1991) Mesocorticolimbic dopaminergic network: functional and regulatory roles. Physiol Rev 71:155-234.

Ledford CC, Fuchs RA, See RE (2003) Potentiated reinstatement of cocaine-seeking behavior following D-amphetamine infusion into the basolateral amygdala. Neuropsychopharmacology 28:1721-1729.

Legault M, Rompre PP, Wise RA (2000) Chemical stimulation of the ventral hippocampus elevates nucleus accumbens dopamine by activating dopaminergic neurons of the ventral tegmental area. J Neurosci 20:1635-1642.

Leslie FM, Loughlin SE, Wang R, Perez L, Lotfipour S, Belluzzia JD (2004) Adolescent development of forebrain stimulant responsiveness: insights from animal studies. Ann N Y Acad Sci 1021:148-159.

Lesniewska B, Miskowiak B, Nowak M, Malendowicz LK (1990) Sex differences in adrenocortical structure and function. XXVII. The effect of ether stress on ACTH and corticosterone in intact, gonadectomized, and testosterone- or estradiol-replaced rats. Res Exp Med (Berl) 190:95-103.

175

Levy AD, Rittenhouse PA, Li Q, Yracheta J, Kunimoto K, Vandekar LD (1994) Influence of repeated cocaine exposure on the endocrine and behavioral-responses to stress in rats. Psychopharmacology 113:547-554.

Li Y, Hu XT, Berney TG, Vartanian AJ, Stine CD, Wolf ME, White FJ (1999) Both glutamate receptor antagonists and prefrontal cortex lesions prevent induction of cocaine sensitization and associated neuroadaptations. Synapse 34:169-180.

Little KY, Kirkman JA, Carroll FI, Clark TB, Duncan GE (1993) Cocaine use increases [3H]WIN 35428 binding sites in human striatum. Brain Res 628:17-25.

Lopez-Rubalcava C, Lucki I (2000) Strain differences in the behavioral effects of antidepressant drugs in the rat forced swimming test. Neuropsychopharmacology 22:191-199.

Loy R, Koziell DA, Lindsey JD, Moore RY (1980) Noradrenergic innervation of the adult rat hippocampal formation. J Comp Neurol 189:699-710.

Macri S, Adriani W, Chiarotti F, Laviola G (2002) Risk taking during exploration of a plus-maze is greater in adolescent than in juvenile or adult mice. Animal Behaviour 64:541-546.

Maisonneuve IM, Ho A, Kreek MJ (1995) Chronic administration of a cocaine "binge" alters basal extracellular levels in male rats: an in vivo microdialysis study. J Pharmacol Exp Ther 272:652-657.

Majchrzak M, Malendowicz LK (1983) Sex differences in adrenocortical structure and function. XII. Stereologic studies of rat adrenal cortex in the course of maturation. Cell Tissue Res 232:457-469.

Mansour A, Meador-Woodruff JH, Bunzow JR, Civelli O, Akil H, Watson SJ (1990) Localization of dopamine D2 receptor mRNA and D1 and D2 receptor binding in the rat brain and pituitary: an in situ hybridization-receptor autoradiographic analysis. J Neurosci 10:2587-2600.

Mantsch JR, Taves S, Khan T, Katz ES, Sajan T, Tang LC, Cullinan WE, Ziegler DR (2007) Restraint-induced corticosterone secretion and hypothalamic CRH mRNA expression are augmented during acute withdrawal from chronic cocaine administration. Neuroscience Letters 415:269-273.

Markou A, Hauger RL, Koob GF (1992) Desmethylimipramine attenuates cocaine withdrawal in rats. Psychopharmacology (Berl) 109:305-314.

Martinez-Sanchis S, Aragon C, Salvador A (2002) Cocaine-induced locomotor activity is enhanced by exogenous testosterone. Physiol Behav 76:605-609.

Matsumoto M, Yoshioka M, Togashi H, Ikeda T, Saito H (1996) Functional regulation by dopamine receptors of serotonin release from the rat hippocampus: in vivo microdialysis study. Naunyn Schmiedebergs Arch Pharmacol 353:621-629.

McClamrock HD, Bass KM, Adashi EY (1991) Ovarian hyperandrogenism: the role of and sensitivity to gonadotropins. Fertil Steril 55:73-79.

McCormick CM, Mathews IZ (2007) HPA function in adolescence: role of sex hormones in its regulation and the enduring consequences of exposure to stressors. Pharmacol Biochem Behav 86:220-233.

McFarland K, Lapish CC, Kalivas PW (2003) Prefrontal glutamate release into the core of the nucleus accumbens mediates cocaine-induced reinstatement of drug-seeking behavior. J Neurosci 23:3531-3537.

McGregor A, Roberts DC (1993) Dopaminergic antagonism within the nucleus accumbens or the amygdala produces differential effects on intravenous cocaine self-administration under fixed and progressive ratio schedules of reinforcement. Brain Res 624:245-252.

McMahon LR, Filip M, Cunningham KA (2001) Differential regulation of the mesoaccumbens circuit by serotonin 5-Hydroxytryptamine (5-HT)2A and 5-HT2C receptors. J Neurosci 21:7781-7787.

Meador-Woodruff JH, Mansour A, Healy DJ, Kuehn R, Zhou QY, Bunzow JR, Akil H, Civelli O, Watson SJ, Jr. (1991) Comparison of the distributions of D1 and D2 dopamine receptor mRNAs in rat brain. Neuropsychopharmacology 5:231-242.

176

Mello NK, Mendelson JH, Kelly M, Bowen CA (2000) The effects of cocaine on basal and human chorionic gonadotropin-stimulated ovarian steroid hormones in female rhesus monkeys. J Pharmacol Exp Ther 294:1137-1145.

Mendelson JH, Teoh SK, Mello NK, Ellingboe J, Rhoades E (1992) Acute effects of cocaine on plasma adrenocorticotropic hormone, luteinizing hormone and prolactin levels in cocaine-dependent men. J Pharmacol Exp Ther 263:505-509.

Meng SZ, Ozawa Y, Itoh M, Takashima S (1999) Developmental and age-related changes of dopamine transporter, and dopamine D1 and D2 receptors in human basal ganglia. Brain Res 843:136-144.

Mikkelsen JD, Hay-Schmidt A, Kiss A (2004) Serotonergic stimulation of the rat hypothalamo-pituitary-adrenal axis: interaction between 5-HT1A and 5-HT2A receptors. Ann N Y Acad Sci 1018:65-70.

Millan MJ, Hjorth S, Samanin R, Schreiber R, Jaffard R, De Ladonchamps B, Veiga S, Goument B, Peglion JL, Spedding M, Brocco M (1997) S 15535, a novel benzodioxopiperazine ligand of serotonin (5-HT)1A receptors: II. Modulation of hippocampal serotonin release in relation to potential anxiolytic properties. J Pharmacol Exp Ther 282:148-161.

Mills K, Ansah TA, Ali SF, Mukherjee S, Shockley DC (2007) Augmented behavioral response and enhanced synaptosomal calcium transport induced by repeated cocaine administration are decreased by calcium channel blockers. Life Sci 81:600-608.

Minabe Y, Emori K, Ashby CR, Jr. (1996) The depletion of brain serotonin levels by para-chlorophenylalanine administration significantly alters the activity of midbrain dopamine cells in rats: an extracellular single cell recording study. Synapse 22:46-53.

Molina VA, Wagner JM, Spear LP (1994) The behavioral response to stress is altered in adult rats exposed prenatally to cocaine. Physiol Behav 55:941-945.

Montague DM, Lawler CP, Mailman RB, Gilmore JH (1999) Developmental regulation of the dopamine D1 receptor in human caudate and putamen. Neuropsychopharmacology 21:641-649.

Muller CP, Carey RJ, De Souza Silva MA, Jocham G, Huston JP (2002) Cocaine increases serotonergic activity in the hippocampus and nucleus accumbens in vivo: 5-HT1a-receptor antagonism blocks behavioral but potentiates serotonergic activation. Synapse 45:67-77.

Muller CP, Thonnessen H, Barros M, Tomaz C, Carey RJ, Huston JP (2004) Hippocampus 5-HT(1A)-receptors attenuate cocaine-induced hyperlocomotion and the increase in hippocampal but not nucleus accumbens 5-HT. Hippocampus 14:710-721.

Nakamura S, Ago Y, Hayashi A, Itoh S, Kakuda M, Hashimoto H, Baba A, Matsuda T (2006) Modification of cocaine-induced behavioral and neurochemical effects by serotonin1A receptor agonist/antagonist in mice. Synapse 60:479-484.

Neisewander JL, Baker DA, Fuchs RA, Tran-Nguyen LTL, Palmer A, Marshall JF (2000) Fos protein expression and cocaine-seeking behavior in rats after exposure to a cocaine self-administration environment. J Neurosci 20:798-805.

Nogueira L, Kalivas PW, Lavin A (2006) Long-term neuroadaptations produced by withdrawal from repeated cocaine treatment: role of dopaminergic receptors in modulating cortical excitability. J Neurosci 26:12308-12313.

Norman AW, Litwack G (1998) Hormones, Second Edition. San Diego, California: Academic press.

O'Dell LE, Torres OV, Natividad LA, Tejeda HA (2007) Adolescent nicotine exposure produces less affective measures of withdrawal relative to adult nicotine exposure in male rats. Neurotoxicol Teratol 29:17-22.

Ojeda SR, Dissen GA, Junier MP (1992) Neurotrophic factors and female sexual development. Front Neuroendocrinol 13:120-162.

Olster DH, Ferin M (1987) Corticotropin-releasing hormone inhibits gonadotropin secretion in the ovariectomized rhesus monkey. J Clin Endocrinol Metab 65:262-267.

177

Orozco-Cabal L, Pollandt S, Liu J, Vergara L, Shinnick-Gallagher P, Gallagher JP (2006) A novel rat medial prefrontal cortical slice preparation to investigate synaptic transmission from amygdala to layer V prelimbic pyramidal neurons. J Neurosci Methods 151:148-158.

Paine, Jackman SL, Olmstead MC (2002) Cocaine-induced anxiety: alleviation by diazepam, but not buspirone, dimenhydrinate or diphenhydramine. Behav Pharmacol 13:511-523.

Palacios JM, Camps M, Cortes R, Probst A (1988) Mapping dopamine receptors in the human brain. J Neural Transm Suppl 27:227-235.

Palanza P, Morley-Fletcher S, Laviola G (2001) Novelty seeking in periadolescent mice: sex differences and influence of intrauterine position. Physiol Behav 72:255-262.

Parsons LH, Justice JB, Jr. (1993a) Serotonin and dopamine sensitization in the nucleus accumbens, ventral tegmental area, and dorsal raphe nucleus following repeated cocaine administration. J Neurochem 61:1611-1619.

Parsons LH, Justice JB, Jr. (1993b) Perfusate serotonin increases extracellular dopamine in the nucleus accumbens as measured by in vivo microdialysis. Brain Res 606:195-199.

Parsons LH, Smith AD, Justice JB, Jr. (1991) The in vivo microdialysis recovery of dopamine is altered independently of basal level by 6-hydroxydopamine lesions to the nucleus accumbens. J Neurosci Methods 40:139-147.

Parsons LH, Koob GF, Weiss F (1995) Serotonin dysfunction in the nucleus accumbens of rats during withdrawal after unlimited access to intravenous cocaine. J Pharmacol Exp Ther 274:1182-1191.

Parsons LH, Koob GF, Weiss F (1996) Extracellular serotonin is decreased in the nucleus accumbens during withdrawal from cocaine self-administration. Behav Brain Res 73:225-228.

Pessia M, Jiang ZG, North RA, Johnson SW (1994) Actions of 5-hydroxytryptamine on ventral tegmental area neurons of the rat in vitro. Brain Res 654:324-330.

Petraglia F, Sutton S, Vale W, Plotsky P (1987) Corticotropin-releasing factor decreases plasma luteinizing hormone levels in female rats by inhibiting gonadotropin-releasing hormone release into hypophysial-portal circulation. Endocrinology 120:1083-1088.

Peyron C, Luppi PH, Kitahama K, Fort P, Hermann DM, Jouvet M (1995) Origin of the dopaminergic innervation of the rat dorsal raphe nucleus. Neuroreport 6:2527-2531.

Phillips PE, Stuber GD, Heien ML, Wightman RM, Carelli RM (2003) Subsecond dopamine release promotes cocaine seeking. Nature 422:614-618.

Pierce RC, Bell K, Duffy P, Kalivas PW (1996) Repeated cocaine augments excitatory amino acid transmission in the nucleus accumbens only in rats having developed behavioral sensitization. J Neurosci 16:1550-1560.

Pierce RC, Reeder DC, Hicks J, Morgan ZR, Kalivas PW (1998) Ibotenic acid lesions of the dorsal prefrontal cortex disrupt the expression of behavioral sensitization to cocaine. Neuroscience 82:1103-1114.

Pitts DK, Marwah J (1987) Cocaine modulation of central monoaminergic neurotransmission. Pharmacol Biochem Behav 26:453-461.

Post RM, Lockfeld A, Squillace KM, Contel NR (1981) Drug-environment interaction: context dependency of cocaine-induced behavioral sensitization. Life Sci 28:755-760.

Potter D, Luther M, Eddy C, Siler-Khodr T, King T, Schenken R (1999) Low-dose follicular-phase cocaine administration disrupts menstrual and ovarian cyclicity in rhesus monkeys. J Soc Gynecol Investig 6:88-94.

Prisco S, Pagannone S, Esposito E (1994) Serotonin-dopamine interaction in the rat ventral tegmental area: an electrophysiological study in vivo. J Pharmacol Exp Ther 271:83-90.

Pum M, Carey RJ, Huston JP, Muller CP (2007) Dissociating effects of cocaine and d-amphetamine on dopamine and serotonin in the perirhinal, entorhinal, and prefrontal cortex of freely moving rats. Psychopharmacology 193:375-390.

178

Pum ME, Carey RJ, Huston JP, Muller CP (2008) Role of medial prefrontal, entorhinal, and occipital 5-HT in cocaine-induced place preference and hyperlocomotion: evidence for multiple dissociations. Psychopharmacology (Berl) 201:391-403.

Quinones-Jenab V, Perrotti LI, Fabian SJ, Chin J, Russo SJ, Jenab S (2001) Endocrinological basis of sex differences in cocaine-induced behavioral responses. Ann NY Acad Sci 937:140-171.

Quinones-Jenab V, Minerly AC, Niyomchia T, Akahvan A, Jenab S, Frye C (2008) Progesterone and allopregnanolone are induced by cocaine in serum and brain tissues of male and female rats. Pharmacol Biochem Behav 89:292-297.

Quinones-Jenab V, Perrotti LI, Ho A, Jenab S, Schlussman SD, Franck J, Kreek MJ (2000) Cocaine affects progesterone plasma levels in female rats. Pharmacol Biochem Behav 66:449-453.

Raap D, Morin B, Medici C, Smith R (2000) Adolescent cocaine and injection stress effects on the estrous cycle. Physiol Behav 70:417-424.

Reid M, Herrera-Marschitz M, Hokfelt T, Terenius L, Ungerstedt U (1988) Differential modulation of striatal dopamine release by intranigral injection of gamma-aminobutyric acid (GABA), dynorphin A and substance P. Eur J Pharmacol 147:411-420.

Reith ME, Li MY, Yan QS (1997) Extracellular dopamine, norepinephrine, and serotonin in the ventral tegmental area and nucleus accumbens of freely moving rats during intracerebral dialysis following systemic administration of cocaine and other uptake blockers. Psychopharmacology (Berl) 134:309-317.

Reith ME, Sershen H, Allen DL, Lajtha A (1983) A portion of [3H]cocaine binding in brain is associated with serotonergic neurons. Mol Pharmacol 23:600-606.

Rivest S, Rivier C (1995) The role of corticotropin-releasing factor and interleukin-1 in the regulation of neurons controlling reproductive functions. Endocr Rev 16:177-199.

Rivier C (1993) Female rats release more corticosterone than males in response to alcohol: influence of circulating sex steroids and possible consequences for blood alcohol levels. Alcohol Clin Exp Res 17:854-859.

Rivier C, Vale W (1987) Cocaine stimulates adrenocorticotropin (ACTH) secretion through a corticotropin-releasing factor (CRF)-mediated mechanism. Brain Res 422:403-406.

Roberts AC, Wallis JD (2000) Inhibitory control and affective processing in the prefrontal cortex: neuropsychological studies in the common marmoset. Cereb Cortex 10:252-262.

Robertson MW, Leslie CA, Bennett JP, Jr. (1991) Apparent synaptic dopamine deficiency induced by withdrawal from chronic cocaine treatment. Brain Res 538:337-339.

Robinson TE, Berridge KC (2003) Addiction. Annu Rev Psychol 54:25-53.

Rocha BA, Fumagalli F, Gainetdinov RR, Jones SR, Ator R, Giros B, Miller GW, Caron MG (1998) Cocaine self-administration in dopamine-transporter knockout mice. Nat Neurosci 1:132-137.

Rogers JL, See RE (2007) Selective inactivation of the ventral hippocampus attenuates cue-induced and cocaine-primed reinstatement of drug-seeking in rats. Neurobiol Learn Mem 87:688-692.

Rosenkranz JA, Grace AA (2002) Cellular mechanisms of infralimbic and prelimbic prefrontal cortical inhibition and dopaminergic modulation of basolateral amygdala neurons in vivo. J Neurosci 22:324-337.

Rossetti ZL, Melis F, Carboni S, Gessa GL (1992) Dramatic depletion of mesolimbic extracellular dopamine after withdrawal from morphine, alcohol or cocaine: a common neurochemical substrate for drug dependence. Ann N Y Acad Sci 654:513-516.

Sarnyai Z, Biro E, Penke B, Telegdy G (1992) The cocaine-induced elevation of plasma corticosterone is mediated by endogenous corticotropin-releasing factor (CRF) in rats. Brain Res 589:154-156.

179

Sarnyai Z, Dhabhar FS, McEwen BS, Kreek MJ (1998) Neuroendocrine-related effects of long-term, 'binge' cocaine administration: Diminished individual differences in stress-induced corticosterone response. Neuroendocrinology 68:334-344.

Sarnyai Z, Mello NK, Mendelson JH, Eros-Sarnyai M, Mercer G (1996) Effects of cocaine on pulsatile activity of hypothalamic-pituitary-adrenal axis in male rhesus monkeys: neuroendocrine and behavioral correlates. J Pharmacol Exp Ther 277:225-234.

Sarnyai Z, Vecsernyés M, Julesz J, Bíró E, Gardi J, Telegdy G (1995) Brain corticotropin-releasing factor mediates 'anxiety-like' behavior induced by cocaine withdrawal in rats. Brain Res 675:89-97.

Satel SL, Price LH, Palumbo JM, McDougle CJ, Krystal JH, Gawin F, Charney DS, Heninger GR, Kleber HD (1991) Clinical phenomenology and neurobiology of cocaine abstinence: a prospective inpatient study. Am J Psychiatry 148:1712-1716.

Scatton B, Simon H, Le Moal M, Bischoff S (1980) Origin of dopaminergic innervation of the rat hippocampal formation. Neurosci Lett 18:125-131.

Schoenbaum G, Roesch MR, Stalnaker TA (2006) Orbitofrontal cortex, decision-making and drug addiction. Trends Neurosci 29:116-124.

Schramm-Sapyta NL, Morris RW, Kuhn CM (2006) Adolescent rats are protected from the conditioned aversive properties of cocaine and lithium chloride. Pharmacology Biochemistry and Behavior 84:344-352.

Schultz W, Dayan P, Montague PR (1997) A neural substrate of prediction and reward. Science 275:1593-1599.

Scibilia RJ, Lachowicz JE, Kilts CD (1992) Topographic nonoverlapping distribution of D1 and D2 dopamine receptors in the amygdaloid nuclear complex of the rat brain. Synapse 11:146-154.

See RE, Kruzich PJ, Grimm JW (2001) Dopamine, but not glutamate, receptor blockade in the basolateral amygdala attenuates conditioned reward in a rat model of relapse to cocaine-seeking behavior. Psychopharmacology (Berl) 154:301-310.

Sesack SR, Bunney BS (1989) Pharmacological characterization of the receptor mediating electrophysiological responses to dopamine in the rat medial prefrontal cortex: a microiontophoretic study. J Pharmacol Exp Ther 248:1323-1333.

Shaham Y, Erb S, Stewart J (2000) Stress-induced relapse to heroin and cocaine seeking in rats: a review. Brain Research Reviews 33:13-33.

Shalev U, Grimm JW, Shaham Y (2002) Neurobiology of relapse to heroin and cocaine seeking: A review. Pharmacol Rev 54:1-42.

Sivam SP (1989) Cocaine selectively increases striatonigral dynorphin levels by a dopaminergic mechanism. J Pharmacol Exp Ther 250:818-824.

Smelson DA, Roy A, Santana S, Engelhart C (1999) Neuropsychological deficits in withdrawn cocaine-dependent males. Am J Drug Alcohol Abuse 25:377-381.

Sobrian SK, Marr L, Ressman K (2003) Prenatal cocaine and/or nicotine exposure produces depression and anxiety in aging rats. Progress in Neuro-Psychopharmacology & Biological Psychiatry 27:501-518.

Sorg BA, Davidson DL, Kalivas PW, Prasad BM (1997) Repeated daily cocaine alters subsequent cocaine-induced increase of extracellular dopamine in the medial prefrontal cortex. J Pharmacol Exp Ther 281:54-61.

Spanagel R, Weiss F (1999) The dopamine hypothesis of reward: past and current status. Trends Neurosci 22:521-527.

Spangler R, Unterwald EM, Kreek MJ (1993) 'Binge' cocaine administration induces a sustained increase of prodynorphin mRNA in rat caudate-putamen. Brain Res Mol Brain Res 19:323-327.

180

Spangler R, Zhou Y, Schlussman SD, Ho A, Kreek MJ (1997) Behavioral stereotypies induced by "binge' cocaine administration are independent of drug-induced increases in corticosterone levels. Behav Brain Res 86:201-204.

Spear L (2000) The adolescent brain and age-related behavioral manifestations. Neurosci Biobehav Rev 24:417-463.

Spear LP (2007) Assessment of adolescent neurotoxicity: rationale and methodological considerations. Neurotoxicol Teratol 29:1-9.

Spear LP, Brake SC (1983) Periadolescence: age-dependent behavior and psychopharmacological responsivity in rats. Dev Psychobiol 16:83-109.

Spector S, Tarver J, Berkowitz B (1972) Effects of drugs and physiological factors in the disposition of catecholamines in blood vessels. Pharmacol Rev 24:191-202.

Stansfield KH, Kirstein CL (2005) Neurochemical effects of cocaine in adolescence compared to adulthood. Brain Res Dev Brain Res 159:119-125.

Summavielle T, Alves CJ, Monteiro PR, Tavares MA (2004) Abnormal immunoreactivity to serotonin in cerebellar Purkinje cells after neonatal cocaine exposure. Ann N Y Acad Sci 1025:630-637.

Suzuki M, Hurd YL, Sokoloff P, Schwartz JC, Sedvall G (1998) D3 dopamine receptor mRNA is widely expressed in the human brain. Brain Res 779:58-74.

Takeuchi Y, Kimura H, Sano Y (1982) Immunohistochemical demonstration of serotonin-containing nerve fibers in the cerebellum. Cell Tissue Res 226:1-12.

Tarazi FI, Tomasini EC, Baldessarini RJ (1998) Postnatal development of dopamine D4-like receptors in rat forebrain regions: comparison with D2-like receptors. Brain Res Dev Brain Res 110:227-233.

Tarazi FI, Tomasini EC, Baldessarini RJ (1999) Postnatal development of dopamine D1-like receptors in rat cortical and striatolimbic brain regions: An autoradiographic study. Dev Neurosci 21:43-49.

Taylor DP, Riblet LA, Stanton HC, Eison AS, Eison MS, Temple DL, Jr. (1982) Dopamine and antianxiety activity. Pharmacol Biochem Behav 17 Suppl 1:25-35.

Teicher MH, Andersen SL, Hostetter JC, Jr. (1995) Evidence for dopamine receptor pruning between adolescence and adulthood in striatum but not nucleus accumbens. Brain Res Dev Brain Res 89:167-172.

Teoh SK, Sarnyai Z, Mendelson JH, Mello NK, Springer SA, Sholar JW, Wapler M, Kuehnle JC, Gelles H (1994) Cocaine effects on pulsatile secretion of ACTH in men. J Pharmacol Exp Ther 270:1134-1138.

Thompson AM, Gosnell BA, Wagner JJ (2002) Enhancement of long-term potentiation in the rat hippocampus following cocaine exposure. Neuropharmacology 42:1039-1042.

Thompson AM, Swant J, Wagner JJ (2005) Cocaine-induced modulation of long-term potentiation in the CA1 region of rat hippocampus. Neuropharmacology 49:185-194.

Thorre K, Ebinger G, Michotte Y (1998) 5-HT4 receptor involvement in the serotonin-enhanced dopamine efflux from the substantia nigra of the freely moving rat: a microdialysis study. Brain Res 796:117-124.

Tirelli E, Laviola G, Adriani W (2003) Ontogenesis of behavioral sensitization and conditioned place preference induced by psychostimulants in laboratory rodents. Neurosci Biobehav Rev 27:163-178.

Todd CL, Grace AA (1999) Modulation of ventral tegmental area dopamine cell activity by the ventral subiculum and entorhinal cortex. Ann N Y Acad Sci 877:688-690.

Trent F, Tepper JM (1991) Dorsal raphe stimulation modifies striatal-evoked antidromic invasion of nigral dopaminergic neurons in vivo. Exp Brain Res 84:620-630.

Trulson ME, Ulissey MJ (1987) Chronic cocaine administration decreases dopamine synthesis rate and increases [3H] spiroperidol binding in rat brain. Brain Res Bull 19:35-38.

181

Tsukada H, Kreuter J, Maggos CE, Unterwald EM, Kakiuchi T, Nishiyama S, Futatsubashi M, Kreek MJ (1996) Effects of binge pattern cocaine administration on dopamine D1 and D2 receptors in the rat brain: An in vivo study using positron emission tomography. J Neurosci 16:7670-7677.

Tzschentke TM, Schmidt WJ (1999) Functional heterogeneity of the rat medial prefrontal cortex: effects of discrete subarea-specific lesions on drug-induced conditioned place preference and behavioural sensitization. Eur J Neurosci 11:4099-4109.

van Megen HJ, Westenberg HG, den Boer JA, Slaap B, Scheepmakers A (1997) Effect of the selective serotonin reuptake inhibitor fluvoxamine on CCK-4 induced panic attacks. Psychopharmacology (Berl) 129:357-364.

Vanderschuren LJ, Di Ciano P, Everitt BJ (2005) Involvement of the dorsal striatum in cue-controlled cocaine seeking. J Neurosci 25:8665-8670.

Volkow ND, Wang G-J, Telang F, Fowler JS, Logan J, Childress A-R, Jayne M, Ma Y, Wong C (2006) Cocaine cues and dopamine in dorsal striatum: Mechanism of craving in cocaine addiction. J Neurosci 26:6583-6588.

Walker QD, Kuhn CM (2008) Cocaine increases stimulated dopamine release more in periadolescent than adult rats. Neurotoxicol Teratol 30:412-418.

Walker QD, Francis R, Cabassa J, Kuhn CM (2001) Effect of ovarian hormones and estrous cycle on stimulation of the hypothalamo-pituitary-adrenal axis by cocaine. J Pharmacol Exp Ther 297:291-298.

Wang GJ, Volkow ND, Fowler JS, Cervany P, Hitzemann RJ, Pappas NR, Wong CT, Felder C (1999) Regional brain metabolic activation during craving elicited by recall of previous drug experiences. Life Sci 64:775-784.

Watanabe H (1984) Activation of dopamine synthesis in mesolimbic dopamine neurons by immobilization stress in the rat. Neuropharmacology 23:1335-1338.

Weddington WW, Brown BS, Haertzen CA, Cone EJ, Dax EM, Herning RI, Michaelson BS (1990) Changes in mood, craving, and sleep during short-term abstinence reported by male cocaine addicts. A controlled, residential study. Arch Gen Psychiatry 47:861-868.

Weiss F, Porrino LJ (2002) Behavioral neurobiology of alcohol addiction: recent advances and challenges. J Neurosci 22:3332-3337.

Weiss F, Maldonado-Vlaar CS, Parsons LH, Kerr TM, Smith DL, Ben-Shahar O (2000) Control of cocaine-seeking behavior by drug-associated stimuli in rats: effects on recovery of extinguished operant-responding and extracellular dopamine levels in amygdala and nucleus accumbens. Proc Natl Acad Sci U S A 97:4321-4326.

Weiss JM, Stojilkovic SS, Diedrich K, Ortmann O (2007) Effects of testosterone on hormonal content and calcium-dependent basal secretion in female rat pituitary cells. J Steroid Biochem Mol Biol 103:149-157.

Williams J, Davies JA (1983) The involvement of 5-hydroxytryptamine in the release of dendritic dopamine from slices of rat substantia nigra. J Pharm Pharmacol 35:734-737.

Williams JM, Steketee JD (2005a) Effects of repeated cocaine on the release and clearance of dopamine within the rat medial prefrontal cortex. Synapse 55:98-109.

Williams JM, Steketee JD (2005b) Time-dependent effects of repeated cocaine administration on dopamine transmission in the medial prefrontal cortex. Neuropharmacology 48:51-61.

Wilson JM, Nobrega JN, Corrigall WA, Coen KM, Shannak K, Kish SJ (1994) Amygdala dopamine levels are markedly elevated after self- but not passive-administration of cocaine. Brain Res 668:39-45.

Winston JS, Gottfried JA, Kilner JM, Dolan RJ (2005) Integrated neural representations of odor intensity and affective valence in human amygdala. J Neurosci 25:8903-8907.

Wise RA, Rompre PP (1989) Brain dopamine and reward. Annu Rev Psychol 40:191-225.

182

Wolf ME, Sun X, Mangiavacchi S, Chao SZ (2004) Psychomotor stimulants and neuronal plasticity. Neuropharmacology 47 Suppl 1:61-79.

Wolf ME, Dahlin SL, Hu XT, Xue CJ, White K (1995) Effects of lesions of prefrontal cortex, amygdala, or fornix on behavioral sensitization to amphetamine: comparison with N-methyl-D-aspartate antagonists. Neuroscience 69:417-439.

Wong DF, Kuwabara H, Schretlen DJ, Bonson KR, Zhou Y, Nandi A, Brasic JR, Kimes AS, Maris MA, Kumar A, Contoreggi C, Links J, Ernst M, Rousset O, Zukin S, Grace AA, Lee JS, Rohde C, Jasinski DR, Gjedde A, London ED (2006) Increased occupancy of dopamine receptors in human striatum during cue-elicited cocaine craving. Neuropsychopharmacology 31:2716-2727.

Yang CR, Mogenson GJ (1984) Electrophysiological responses of neurones in the nucleus accumbens to hippocampal stimulation and the attenuation of the excitatory responses by the mesolimbic dopaminergic system. Brain Res 324:69-84.

Yoshimura S, Sakamoto S, Kudo H, Sassa S, Kumai A, Okamoto R (2003) Sex-differences in adrenocortical responsiveness during development in rats. Steroids 68:439-445.

Yoshioka M, Matsumoto M, Togashi H, Saito H (1995) Effects of conditioned fear stress on 5-HT release in the rat prefrontal cortex. Pharmacol Biochem Behav 51:515-519.

Zhang Y, Schlussman SD, Ho A, Kreek MJ (2003) Effect of chronic "binge cocaine" on basal levels and cocaine-induced increases of dopamine in the caudate putamen and nucleus accumbens of C57BL/6J and 129/J mice. Synapse 50:191-199.

Zhou Y, Spangler R, Ho A, Jeanne Kreek M (2001) Hypothalamic CRH mRNA levels are differentially modulated by repeated 'binge' cocaine with or without D(1) dopamine receptor blockade. Brain Res Mol Brain Res 94:112-118.

Zhou Y, Spangler R, Schlussman S, Ho A, Kreek M (2003) Alterations in hypothalamic-pituitary-adrenal axis activity and in levels of proopiomelanocortin and corticotropin-releasing hormone-receptor 1 mRNAs in the pituitary and hypothalamus of the rat during chronic 'binge' cocaine and withdrawal. Brain Res 964:187-199.

Zhou Y, Spangler R, LaForge KS, Maggos CE, Ho A, Kreek MJ (1996) Corticotropin-releasing factor and type 1 corticotropin-releasing factor receptor messenger RNAs in rat brain and pituitary during "binge"-pattern cocaine administration and chronic withdrawal. J Pharmacol Exp Ther 279:351-358.

Zhou Y, Spangler R, Yuferov VP, Schlussmann SD, Ho A, Kreek MJ (2004) Effects of selective D1- or D2-like dopamine receptor antagonists with acute "binge" pattern cocaine on corticotropin-releasing hormone and proopiomelanocortin mRNA levels in the hypothalamus. Brain Res Mol Brain Res 130:61-67.

Zola-Morgan S, Squire LR, Alvarez-Royo P, Clower RP (1991) Independence of memory functions and emotional behavior: separate contributions of the hippocampal formation and the amygdala. Hippocampus 1:207-220.

Zorrilla E, Valdez G, Weiss F (2001) Changes in levels of regional CRF-like-immunoreactivity and plasma corticosterone during protracted drug withdrawal in dependent rats. Psychopharmacology (Berl) 158:374-381.

183

Chapter 4

Long-term Effects of Cocaine throughout Adolescence in the Rat

4.1. Introduction

Long-lasting effects of cocaine exposure have already been reported in an

extensive body of literature. There are numerous reports of long-lasting neurobiological

and behavioural consequences of the prenatal cocaine exposure, which last to adult life.

For example, prenatal cocaine exposure is reported to produce depression and anxiety in

aging rats (for reviews, see Sobrian et al., 2003), to produce alterations on aggressive

behaviour in adult rats (Johns et al., 1994), and to result in increased rewarding effects of

cocaine in the adult mice (Malanga et al., 2008). In adult animals, long-term

neurobiological and behavioural abnormalities induced by cocaine exposure, such as

dopaminergic neuroadaptations (Ungless et al., 2001; Kimmel et al., 2003; Nogueira et al.,

2006) and enhanced levels of anxiety behaviour (Erb et al., 2006), were also reported.

During adolescence, the developing brain may be more vulnerable to long-lasting

modifications by drug exposure (Andersen, 2003). In fact, a major concern in adolescent

psychostimulant abuse is the long-term consequence of this practice, since early drug

exposure may induce long-term adaptations, which, among other effects, may render the

organism more susceptible to drug abuse later in life (Spear, 2000). In fact, epidemiologic

data showed that drug use in adolescence is a reliable predictor of drug abuse in

adulthood (Merline et al., 2004). Studies in rodents show that early developmental

exposure to psychostimulants causes behavioural alterations in sensitivity to drugs of

abuse and natural rewards that endure into adulthood (Andersen et al., 2002; Bolanos et

al., 2003), raising the possibility that early exposure to psychostimulants can have long-

term negative consequences (Carlezon and Konradi, 2004). However, in some studies,

the adolescent brain appears resilient, showing few long-term effects of substance abuse

(Santucci et al., 2004).

The propose to determine whether adolescence represents a window of

vulnerability to the deleterious effects of cocaine or a period of increased resilience to the

effects of cocaine leading to weak long-lasting effects, represents a current and very

important issue of investigation.

The work described in this chapter was performed in order to evaluate the long-

term effects of chronic cocaine exposure during the adolescence period upon the activity

of the HPA and the HPG axes of both male and female wistar rats, by assessing the

hormonal plasma levels 5 and 10 days after cocaine withdrawal. The effects of cocaine

exposure were also evaluated upon the dopaminergic and serotonergic systems by

assessing the monoamines levels, in areas known to be involved with drug addiction, also

5 and 10 days after cocaine withdrawal.

187

The discontinuation of cocaine use leads to the expression of various withdrawal

symptoms, including depression, anxiety, and craving (Gawin and Kleber, 1986;

Weddington et al., 1990; Satel et al., 1991; Coffey et al., 2000). Moreover, these

symptoms provide part of the negative reinforcement associated with cocaine

dependence and are important motivational factors for relapse and maintenance of

repetitive cycles of cocaine abuse (Gawin et al., 1989; Shaham et al., 2000; Shalev et al.,

2002). Thus, it was defined the objective of evaluate the effects of chronic cocaine

exposure during adolescence period, on behavioural strategies adopted to cope with an

environmental anxiogenic situation during withdrawal period, by submitting the animals 5

and 10 days after withdrawal to the EPM.

Psychostimulant drugs such as cocaine also have profound and long-lasting

effects on social behaviours (Wood and Spear, 1998; Estelles et al., 2005). Thus,

evaluate the long-term effects of cocaine administration during adolescence on the

aggressive behaviour, was also defined as an objective by testing male rats 10 days after

withdrawal in the resident-intruder paradigm.

188

4.2. Material and Methods

4.2.1. Animals

Using the animal paradigm described in Chapter 2, we have obtained animals

chronically exposed to cocaine during adolescence that were used in the following

experiments. Briefly, on PND 30 animals were housed individually, and were randomly

assigned to the different experimental groups. Only one male and one female from each

litter were used in each experimental group. Rats were treated following a binge pattern

administration of cocaine, each day being injected 3 times intraperitonealy (i.p.) with

15mg/kg of body weight cocaine hydrochloride dissolved in 0.9% NaCl solution from PND

35 to PND 50. The daily injections were administered at hourly intervals, between 9:00

and 11:00 a.m., in a volume of 1 mL/kg. Controls were given an isovolumetric 0.9% NaCl

solution following the same experimental protocol. During the entire period of drug

administration animals were daily weighed. At least 6 animals were used in each

experimental group.

4.2.2. Behavioural tests

4.2.2.1. Elevated plus maze test

On PND 55 and on PND 60, 5 and 10 days after withdrawal, male Wistar rats were

tested in an EPM apparatus as described in Chapter 2. Spatiotemporal and behavioural

parameters were registered at the time of behaviour analyses. Spatiotemporal measures

comprised the frequencies of total, open and closed arm entries and central platform

entries, as well as the time spent in the open, closed and central part of the maze.

Behavioural measures comprised frequency and duration scores for rearing, head dipping

and grooming.

4.2.2.2. Resident-intruder paradigm

On PND 60, 10 days after cocaine withdrawal, aggressive behaviour of male rats

was evaluated using the resident-intruder paradigm, as described in Chapter 2.

Behavioural activity during test was analysed, and frequency, duration and latency data

were collected for the following behaviours divided in to three categories: (1) offensive

behaviours that included attack, bite, pinning, wrestling, chasing and aggressive

grooming; (2) defensive behaviours that included flight, avoidance of contact and

189

submissive-supine posture; (3) social investigation behaviours including sniffing,

anogenital sniffing, and approach behaviour.

4.2.3. Blood and tissue collection

Five and 10 day after the last administration of cocaine, and after the behavioural

tests, animals were sacrificed by decapitation and trunk blood was collected in lithium

heparin cover tubes and centrifuged at 1600g for 15 minutes; the obtained plasma was

kept at -70ºC until hormonal assays were conducted. Brains were also rapidly removed

and SN-VTA, Nac, dorsal striatum, prefrontal cortex, amygdala, hippocampus, dorsal

raphe nucleus, cerebellum and a hypothalamic section containing the suprachiasmatic,

pre-optic and paraventricular nuclei were dissected on ice. Pituitary glands were also

collected. Tissue samples were frozen by immersion in 2-methylbutane cooled over dry

ice and stored at -70ºC until used for further analyses. Five and 10 day after the last

administration of cocaine gonads and adrenal glands were collected and weighed, and the

data obtained was registered.

4.2.4. Hormone plasma levels determinations

The levels of progesterone, estradiol, testosterone, corticosterone, and ACTH,

were measured on plasma obtained 5 and 10 days after withdrawal. The levels of

corticosterone were also measured on the plasma obtained after the behavioural tests. All

hormones plasma levels were measured by enzyme immunoassay, using commercially

available assays.

The progesterone, estradiol, testosterone and corticosterone plasma levels were

measured using assays provided by Cayman Chemical Company (Ann Arbor, MI, USA).

For progesterone, the assay sensitivity was 0.012 ng/mL, and the intra- and inter-assay

coefficients of variation were less 8% and 12%, respectively. For estradiol, the assay

sensitivity was 0.004 ng/mL, and the intra- and inter-assay coefficients of variation were

less 10% and 12%, respectively. For testosterone, the assay sensitivity was 0.004 ng/mL,

and the intra- and inter-assay coefficients of variation were less 10% and 15%,

respectively. For corticosterone, the assay sensitivity was 0.04 ng/mL, and the intra- and

inter-assay coefficients of variation were less than 7% and 9%, respectively. The ACTH

plasma levels were measured using an assay provided by Phoenix Pharmaceuticals, Inc.

(Belmont, California, USA). The ACTH assay sensitivity was 0.18 ng/mL, and the intra-

and inter-assay coefficients of variation were less than 5 % and 14%, respectively.

190

Corticosterone plasma levels after the behavioural tests were measured using a kit

provided by Immunodiagnostic Systems Ltd (Boldon, UK). Assay sensitivity was 0.23

ng/mL. The intra- and inter-assay coefficients of variation were less than 3.1% and 7.2%,

respectively.

4.2.5. Neurochemical determinations

Concentrations of DA and 5-HT and their metabolites, 5-HIAA, DOPAC, and HVA

were quantified by HPLC-EC, in the dissected brain areas (SN-VTA, Nac, dorsal striatum,

prefrontal cortex, amygdala, hippocampus, dorsal raphe nucleus, cerebellum and

hypothalamus) collected 5 and 10 days after withdrawal, as described in Chapter 2. After

each behavioural test, levels of DA and 5-HT and their metabolites were determined in the

SN-VTA, dorsal raphe nucleus, amygdala, hippocampus, prefrontal cortex and

hypothalamus. Concentrations are expressed in ng/mg protein. The ratios of DOPAC to

DA, HVA to DA and DOPAC and HVA to DA were determined for each animal and used

as indexes of DA turnover rate, whereas the ratio of 5-HIAA to 5-HT was used as an index

of 5-HT turnover rate.

4.2.6. mRNA expression levels determination

The expression levels of mRNA transcripts for tyrosine hydroxylase (TH) were

measured in SN-VTA, Nac and dorsal striatum, and the expression levels of mRNA

transcripts for monoamine oxidase-A (MOA-A), DA transporter (DAT), DA receptors (D1,

D2, D3) and GR were measured in the SN-VTA, Nac, dorsal striatum, prefrontal cortex,

amygdala, hippocampus and hypothalamus, brain areas dissected from males sacrificed

10 days after last the administration, by qRT-PCR, as described in Chapter 2. Additionally,

the expression levels of mRNA transcripts for CRH and androgen receptor (AR) were

measured in the hypothalamus, and the expression levels of mRNA transcripts for CRH

receptor 1 (CRH-R1), GnRH receptor (GnRH-R), AR, LHβ, GR and proopiomelanocortin

(POMC) were measure in the pituitary.

The reference gene, glyceraldahyde-3-phosphate-dehydrogenase (GAPDH) was

used as internal standard for normalization.

Primer sequences and annealing temperatures (Ta) for each gene are presented

in table 4.2.2.4.1 and table 4.2.2.4.2.

191

Table 4.2.2.4.1- Primer pairs and their sequences, PCR segment size and Ta used in the qRT-PCR to study the expression of dopaminergic- related mRNAs.

Gene Primer sequence Size (bp) Tm (ºC) Ta (ºC)

GAPDH Sense: 5’-ttc aac ggc aca gtc aag g-3’

Antisense: 5’-ctc agc acc agc atc acc-3’ 114 75.9 55

TH Sense: 5’-ggc ttc tct gac cag gtg tat c-3’

Antisense: 5’-caa tct ctt ccg ctg tgt att cc-3’ 112 75.5 55

DAT Sense: 5’-gtc att gtt ctg ctc tac ttc-3’

Antisense: 5’-gtc cac act gag gta tgc-3’ 165 78.2 55

MAO-A Sense: 5’-ggc aca gag aca gca aca c-3’

Antisense: 5’-cag acc agg cac gga agg-3’ 204 77.5 59

D1 Sense: 5’-act ctg tct gtc ctt ata tcc ttc-3’

Antisense: 5’-gtt gtc atc ctc ggt gtc c-3’ 114 75.2 55

D2 Sense: 5’-caa caa tac aga cca gaa tgc gtg-3’

Antisense: 5’-cag cag agt gac gat gaa gg-3’ 100 72.8 55

D3 Sense: 5’-gtc cgc acg cct act acg-3’

Antisense: 5’-cag cac agc agc aca tac c-3’ 86 75.9 55

GR Sense: 5’-gga cag cct gac ttc ctt gg-3’

Antisense: 5’-tcc agg gct tga gta ccc at-3’ 76 74.2 55

D1- dopamine receptor 1; D2- dopamine receptor 2; D3- dopamine receptor 3; DAT- dopamine transporter; GAPDH- Glyceraldahyde-3-phosphate-dehydrogenase; GR- Glucocorticoid receptor; MAO-A- Monoamine oxidase A; TH- Tyrosine hydroxilase; Ta- anneling temperature; Tm- melting temperature.

192

Table 4.2.2.4.2- Primer pairs and their sequences, PCR segment size and Ta used in the qRT-PCR to study the the expression of HPA and HPG axes- related mRNAs.

Gene Primer sequence Size (bp) Tm (ºC) Ta (ºC)

GAPDH Sense: 5’-ttc aac ggc aca gtc aag g-3’

Antisense: 5’-ctc agc acc agc atc acc-3’ 114 75.9 55

CRH Sense: 5’-gga gaa gag aaa gga gaa gag g-3’

Antisense: 5’-aga atc ggc tga ggt tgc tg-3’ 283 83.3 62

CRH-R1 Sense: 5’-ctt ctt ctg gat gtt cgg tga g-3’

Antisense: 5’-atg agg atg cgg aca atg ttg-3’ 279 78.9 59

GnRH-R Sense: 5’-aac atc agc agt aac aga gac-3’

Antisense: 5’-aga ggc att gaa ggc agt ag-3’ 216 76.9 55

AR Sense: 5’-aag cca ttg agc cag gag tg-3’

Antisense: 5’-tta gtg aag gac cgc caa cc-3’ 235 78.5 55

POMC Sense: 5’-gaa gcg gcg ccc tgt gaa-3’

Antisense: 5’-ctc gcc ttc cag ctc cct ctt-3’ 94 78.5 59

LHβ Sense: 5’-cta ctg tcc tag cat ggt tcg-3’

Antisense: 5’-gga agg tca cag gtc att gg-3’ 231 82.7 59

GR Sense: 5’-gga cag cct gac ttc ctt gg-3’

Antisense: 5’-tcc agg gct tga gta ccc at-3’ 76 74.2 55

Ar- Androgen receptor; CRH- Corticotrophin-releasing hormone; CRH-R1- Corticotrophin-releasing hormone receptor 1; GAPDH- Glyceraldahyde-3-phosphate-dehydrogenase; GnRH-R- Gonadotropin-releasing hormone receptor; GR- Glucocorticoid receptor; LHβ- Luteinizing hormone β-subunit; POMC- Proopiomelanocortin; Ta- anneling temperature; Tm- melting temperature.

4.2.7. Statistical analysis

A 2-way ANOVA with treatment (cocaine or saline) and sex as between-subject

fixed-factors with repeated measures was used to evaluate body weight evolution during

the experimental period until long-term withdrawal. Student’s t-test was used as a post-

hoc test to analyse statistical significant differences.

Adrenal and gonadal weights were converted in percentage of body weight, and

adrenal data was analyzed by a 3-way ANOVA with time point of assessment (5 and 10

days), treatment and sex as between-subject fixed-factors. Gonadal data was separated

by sex and analyzed by a 2-way ANOVA (time point of assessment (5 and 10 days) x

treatment).

For hormonal and neurochemical data obtained at 5 and 10 days after withdrawal,

obtained after the EPM and after the aggression test, statistical significance between

groups was assessed using the non-parametric Mann-Whitney U Test.

193

Student’s t-test was used to compare the expression levels of the mRNAs,

obtained by qRT-PCR, between groups.

The EPM behaviour data was analysed by a 2-way ANOVA with treatment

(cocaine or saline) and time point of assessment (5 and 10 days after withdrawal) as

between-subject fixed-factors and behavioural measurements as the dependent variable.

Student’s t-test was used as a post-hoc test to analyse statistical significant differences.

Analysis of behaviour data obtained in the aggression test was performed using

the Student’s t-test.

Differences were considered to be statistically significant when p<0.05. All data are

expressed as the mean plus standard error of the mean (S.E.M).

Statistical analyses were performed using the software SPSS 14.0 (SPSS INC. Chicago, Illinois, USA).

194

4.3. Results

4.3.1. Basal analyses

4.3.1.1. Biometric measures

Evolution of body weight. Cocaine-treated and control animals were weight

everyday from PND 35 to PND 50, and on PND 55 and PND 60. Data were analyzed with

a 2-way ANOVA (treatment x sex) with repeated measures. The body weight gain was

statistically different between male and female rats [F(1,20)= 92.787; p<0.001]), post-hoc

comparisons using Student’s t-test confirm that from PND 38 until the end of exposure

period and on PND 55 and PND 60 the weight gain was higher in males than in females

(figure 4.3.1.1.1.). The 2-way ANOVA with repeated measures also revealed that there

were no differences in body weight gain between cocaine-treated males and control males

and between cocaine-treated and control females.

0

50

100

150

200

35 37 39 41 43 45 47 49 55 60

PND

Wei

ght g

ain

(g)

saline male cocaine male saline female cocaine female

Figure 4.3.1.1.1- Effects of chronic cocaine exposure on body weight gain during exposure period and

withdrawal. Values are mean ± S.E.M.. Saline male, n=7; cocaine male, n=7; saline female, n=5; cocaine

female, n=5. PND- postnatal day. &, sex effect; p<0.001.

Adrenal weight. Adrenals were collected 5 and 10 days after cocaine withdrawal

and their weights were recorded. The weight data was converted in percentage of body

weight and were analyzed with a 3-way ANOVA (time point of assessment- 5 or 10 days x

treatment x sex). The statistical analysis revealed a main effect of sex in the adrenal

weight [F(1,46)=97.731; p<0.001] (figure 4.3.1.1.2). The adrenal glands weight in females

was higher than in males. No treatment effects were found in males or females. The

adrenal weight also did not differ between 5 and 10 days after cocaine withdrawal.

&

/ /

195

Adrenal glands

0

0.02

0.04

0.06

5 days 10 days

% o

f bod

y w

eigh

t

saline male cocaine male saline female cocaine female

Figure 4.3.1.1.2- Effects of chronic cocaine exposure throughout adolescence on adrenal weight of male and

female rats assessed 5 and 10 days after the withdrawal. Each column represents the mean + S.E.M. for a

least 6 animals per group.

Gonadal weight. Testes and ovaries were collected 5 and 10 days after withdrawal

of cocaine and their weight were recorded. The percentage of body weight was calculated

and data was analyzed by a 2-way ANOVA (age x treatment). In male group a 2-way

ANOVA revealed an interaction between time point of assessment (5 and 10 days) x

treatment [F(1,24)=4.306; p<0.05], a post-hoc analysis using Student’s t-test indicate that

at 10 days after withdrawal the testes weight were lower in cocaine-treated males as

compared to saline males (t(10)=-2.913; p<0.02) (figure 4.3.1.1.3-A). The statistical

analysis revealed no significant effects of cocaine treatment in the weight of female

gonads, both when assessed 5 or 10 days after withdrawal (figure 4.3.1.1.3-B). Also, no

differences were observed between time points of assessment in female gonadal weight.

196

A Male gonads B Female gonads

0

0.4

0.8

1.2

5 days 10 days

% o

f bod

y w

eigh

t

saline male cocaine male

00.020.040.060.08

5 days 10 days

% o

f bod

y w

eigh

t

saline female cocaine female

Figure 4.3.1.1.3- Effects of chronic cocaine exposure throughout adolescence on (A) male gonads and (B)

females gonads weights assessed 5 and 10 days after cocaine withdrawal. Each column represents the

mean + S.E.M. for at least 6 animals per group. Columns with the same letter are significantly different from

each other. a p< 0.02.

4.3.1.2. Hormone plasma levels

The levels of hormones were measured on plasma obtained 5 and 10 days after

cocaine withdrawal. The data was analyzed by the non-parametric Mann-Whitney U Test.

ACTH. The plasma levels of ACTH were not significant different between cocaine-

treated males and females, and their respective controls, in the two assessment time

points (figure 4.1.2.1-A). In saline female group, the levels of ACTH increase from 5 to 10

days after the last administration [Z=-2.000 (2-tailed) p<0.05], this effect was not observed

in saline male group. Cocaine-treated males also presented an increase in levels of ACTH

from 5 days to 10 days that almost reach significance [Z=-1.939 (2-tailed) p=0.053].

Corticosterone. No significant effects of cocaine treatment were found in both

male and female rats, at the two time point assessment (figure 4.1.2.1-B).

Testosterone. In the male group, 10 days after withdrawal cocaine-treated males

presented lower levels of testosterone [Z=-2.082 (2-tailed) p<0.05]. Saline-treated males

displayed an increase in testosterone levels from 5 to 10 days after the last administration

[Z=-2.562 (2-tailed) p<0.02], this effect was not present in cocaine-treated males. No

significant differences were observed in testosterone levels between cocaine-treated and

saline-treated females at both time points of assessment (figure 4.3.1.2.2-B).

Progesterone. In the saline-treated females the levels of progesterone decreased

from 5 to 10 days after the last administration [Z=-2.242 (2-tailed) p<0.05]. This effect was

not observed in cocaine-treated females. In fact, 10 days after withdrawal, cocaine-treated

females presented increased levels of progesterone as compared to saline females [Z=-

2.166 (2-tailed) p<0.05]. No significant differences were found in the plasma levels of

a a

197

progesterone between cocaine- and saline-treated males at the both time point

assessment (figure 4.3.1.2.2- C).

Estradiol. In the saline-treated female group, the levels of estradiol increased

from 5 to 10 days after the end of the administration period [Z=-2.571 (2-tailed) p<0.02]

(figure 4.3.1.1.2-E). This effect was not observed in cocaine-treated females.

A

0

2

4

6

8

5 days 10 days

AC

TH le

vels

(ng/

mL)

saline male cocaine male saline female cocaine female

B

0

20

40

60

80

100

5 days 10 days

Cor

ticos

tero

ne le

vels

(ng/

mL)

saline male cocaine male saline female cocaine female

Figure 4.3.1.2.1- Effects of chronic cocaine exposure throughout adolescence on (A) ACTH and (B)

corticosterone plasma levels assessed 5 and 10 days after withdrawal. Each column represents the mean +

S.E.M. for at least 5 animals per group. ACTH- Adrenocorticotropic hormone. Columns with the same letter

are significantly different from each other, a p<0.05.

a

a

198

A B

01234

5 days 10 daysTest

oste

rone

leve

ls

(ng/

mL)

saline male cocaine male

0.00

0.05

0.10

0.15

5 days 10 daysTest

oste

rone

leve

ls

(ng/

mL)

saline female cocaine female

C D

0

0.2

0.4

0.6

5 days 10 daysProg

este

rone

leve

ls

(ng/

mL)

saline male cocaine male

0

50

100

150

5 days 10 daysProg

este

rone

leve

ls

(ng/

mL)

saline female cocaine female

E

0

15

30

45

5 days 10 days

Estra

diol

leve

ls

(ng/

mL)

saline female cocaine female

Figure 4.3.1.2.2- Effects of chronic cocaine exposure throughout adolescence on plasma levels of

testosterone in (A) males and (B) females, progesterone in (C) males and (D) females and estradiol in (E)

females, assessed 5 and 10 days after cocaine withdrawal. Each column represents the mean + S.E.M. for at

least 5 animals per group. Columns with the same letter are significantly different from each other. a,e p<0.02, b,c,d p<0.05.

4.3.1.3. Neurochemical determinations

The levels of DA, 5-HT and their metabolites, DOPAC, HVA and 5-HIAA were

measured, by HPLC-EC, 5 and 10 days after the end of the exposure period, in the

following brain regions: SN-VTA, Nac, dorsal striatum, prefrontal cortex, hippocampus,

amygdala, hypothalamus, dorsal raphe nucleus and cerebellum. These different brain

areas were selected based on their involvement in cocaine response. The data obtained

were analyzed by the non-parametric Mann-Whitney U Test.

a,b

b a

c,d

d

c

e

e

199

Within the SN-VTA, statistical analysis revealed that 10 days after withdrawal,

cocaine-treated males displayed higher levels of DOPAC as compared to saline males

and cocaine females ([Z=-2.082 (2-tailed) p<0.037] and [Z=-2.429 (2-tailed) p<0.02],

respectively) (figure 4.3.1.3.1). Cocaine-treated males also displayed an increase in 5-HT

levels from 5 to 10 days after the end of treatment [Z=-2.593 (2-tailed) p<0.02]. At 5 days

after withdrawal, no differences were detected in the levels of DA, 5-HT and their

metabolites between cocaine-treated animals and their respective controls, also no

differences were found in DA and 5-HT turnover rates between the experimental groups.

Within the Nac, no differences were detected in the levels of DA and 5-HT and

their metabolites between cocaine-treated animals and their respective controls at both

time points of analysis (figure 4.3.1.3.2). Also, no differences were found in both DA and

5-HT turnover rates between the experimental groups (table 4.3.1.3.2). However, in

cocaine-treated female group the levels of DA, 5-HT and 5-HIAA increased from 5 to 10

days after the end of treatment ([Z=-1.981 (2-tailed) p<0.05], [Z=-2.236 (2-tailed) p<0.05]

and [Z=-2.108 (2-tailed) p<0.05], respectively).

200

SN-VTA

A

0

200

400

600

800

DA DOPAC HVA DA DOPAC HVA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

5 days 10 days

B

0

200

400

600

800

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

5 days 10 days

Figure 4.3.1.3.1- Effects of chronic cocaine exposure throughout adolescence on the (A) levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the SN-VTA,

determined by HPLC-EC for male and female rats, 5 and 10 days after withdrawal. Each column represents

the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for at least 5 animals

per group. Columns with the same letter are significantly different from each other. a p<0.05, b,c p<0.02.

a a,b b

c

c

201

Nucleus accumbens

A

0

1500

3000

4500

6000

DA DOPAC HVA DA DOPAC HVA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

5 days 10 days

B

0

200

400

600

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

5 days 10 days

Figure 4.3.1.3.2- Effects of chronic cocaine exposure throughout adolescence on the (A) levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the Nac, determined

by HPLC-EC for male and female rats, 5 and 10 days after withdrawal. Each column represents the mean +

S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for at least 5 animals per group.

Columns with the same letter are significantly different from each other. a,b,c p<0.05.

a

a

b

b

c

c

202

Table 4.3.1.3.1- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the SN-VTA, 5 and 10 days after the end of cocaine exposure period.

5 days 10 days

Male Female Male Female

saline cocaine saline cocaine saline cocaine saline cocaine

DOPAC/DA 0.16±0.01

(n=8)

0.21±0.02

(n=9)

0.16±0.01

(n=7)

0.23±0.05

(n=7)

0.24±0.06

(n=6)

0.16±0.02

(n=6)

0.16±0.02

(n=5)

0.14±0.01

(n=7)

HVA/DA 0.09±0.02

(n=8)

0.11±0.02

(n=9)

0.07±0.01

(n=7)

0.13±0.03

(n=7)

0.09±0.02

(n=6)

0.07±0.02

(n=6)

0.09±0.02

(n=5)

0.07±0.01

(n=7)

(DOPAC+HVA)/DA 0.25±0.02

(n=8)

0.32±0.03

(n=9)

0.23±0.02

(n=7)

0.36±0.07

(n=7)

0.33±0.05

(n=6)

0.24±0.04

(n=6)

0.25±0.04

(n=5)

0.22±0.02

(n=7)

5-HIAA/5-HT 0.27±0.03

(n=8)

0.24±0.03

(n=9)

0.28±0.08

(n=7)

0.40±0.11

(n=7)

0,28±0.06

(n=6)

0.22±0.03

(n=6)

0.16±0.02

(n=5)

0.57±0.33

(n=7)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. In the same line, values with

same letter are significantly different. DA- dopamine; DOPAC- 3,4-dihydroxyphenylacetic acid; HVA-

homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid.

Table 4.3.1.3.2- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the Nac, at 5 and 10 days after the end of cocaine exposure period.

5 days 10 days

Male Female Male Female

saline cocaine saline cocaine saline cocaine saline cocaine

DOPAC/DA 0.25±0.03

(n=8)

0.28±0.03

(n=9)

0.28±0.03

(n=7)

0.30±0.06

(n=7)

0.28±0.03

(n=6)

0.27±0.03

(n=5)

0.24±0.02

(n=6)

0.25±0.01

(n=7)

HVA/DA 0.07±0.01

(n=8)

0.10±0.01

(n=9)

0.08±0.01

(n=7)

0.11±0.03

(n=7)

0.08±0.01

(n=6)

0.07±0.01

(n=5)

0.06±0.01

(n=6)

0.08±0.01

(n=7)

(DOPAC+HVA)/DA 0.33±0.03

(n=8)

0.38±0.04

(n=9)

0.36±0.03

(n=7)

0.41±0.09

(n=7)

0.36±0.04

(n=6)

0.33±0.03

(n=5)

0.29±0.02

(n=6)

0.33±0.02

(n=7)

5-HIAA/5-HT 0.30±0.14

(n=8)

0.37±0.12

(n=9)

0.30±0.09

(n=7)

0.33±0.12

(n=7)

0.34±0.19

(n=6)

0.59±0.27

(n=5)

0.45±0.16

(n=6)

0.34±0.12

(n=7)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid.

Within the dorsal striatum, 5 days after withdrawal no differences were detected in

the levels of DA, 5-HT and their metabolites between cocaine-treated animals and their

respective controls. Ten days after withdrawal cocaine-treated males presented higher

HVA levels as compared to control males [Z=-2.082 (2-tailed) p<0.05] (figure 4.3.1.3.3). In

saline-treated male groups the levels of HVA decreased from 5 to 10 days after the end of

treatment [Z=-2.082 (2-tailed) p<0.02], moreover, it was also observed a decrease in

HVA/DA turnover rate [Z=-2.082 (2-tailed) p<0.05]. Ten days after the last administration,

203

saline-treated males also presented lower levels of 5-HIAA as compared to saline-treated

females [Z=-2.402 (2-tailed) p<0.02], this effects was not observed in cocaine-treated

animals.

Dorsal striatum

A

0

2000

4000

6000

8000

DA DOPAC HVA DA DOPAC HVA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

5 days 10 days

B

0

200

400

600

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

5 days 10 days

Figure 4.3.1.3.3- Effects of chronic cocaine exposure throughout adolescence on the (A) levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the dorsal striatum,

determined by HPLC-EC for male and female rats, 5 and 10 days after withdrawal. Each column represents

the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for at least 6 animals

per group. Columns with the same letter are significantly different from each other. a p<0.05, b,c p<0.02

a,b b

c

c

a

204

Table 4.3.1.3.3- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the dorsal striatum, 5 and 10 days after the end of cocaine exposure period.

5 days 10 days

Male Female Male Female

saline cocaine saline cocaine saline cocaine saline Cocaine

DOPAC/DA 0.23±0.03

(n=8)

0.21±0.02

(n=8)

0.19±0.03

(n=7)

0.19±0.02

(n=7)

0.21±0.01

(n=6)

0.18±0.02

(n=6)

0.19±0.03

(n=6)

0.19±0.02

(n=7)

HVA/DA 0.11±0.02

(n=8)

0.10±0.02

(n=8)

0.08±0.01

(n=7)

0.09±0.01

(n=7)

0.05±0.01

(n=6)

0.07±0.01

(n=6)

0.06±0.01

(n=6)

0.08±0.01

(n=7)

(DOPAC+HVA)/DA 0.34±0.04

(n=8)

0.31±0.03

(n=8)

0.27±0.03

(n=7)

0.28±0.03

(n=7)

0.26±0.02

(n=6)

0.25±0.03

(n=6)

0.25±0.03

(n=6)

0.26±0.03

(n=7)

5-HIAA/5-HT 0.36±0.17

(n=8)

0.31±0.16

(n=8)

0.25±0.17

(n=7)

0.24±0.14

(n=7)

0.38±0.18

(n=6)

0.36±0.22

(n=6)

0.35±0.19

(n=6)

0.28±0.11

(n=7)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. In the same line, values with

same letter are significantly different. DA- dopamine; DOPAC- 3,4-dihydroxyphenylacetic acid; HVA-

homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid. a p<0.05.

Within the prefrontal cortex, no statistical significant differences were found on the

levels of DA, 5-HT and their metabolites or their turnover between the experimental

groups (figure 4.3.1.3.4 and table 4.3.1.3.4).

Within the hippocampus, the statistical analysis revealed that in cocaine-treated

female group the levels of DA and 5-HIAA increased from 5 to 10 days after cocaine

withdrawal ([Z=-2.082 (2-tailed) p<0.05] and [Z=-2.242 (2-tailed) p<0.05], respectively)

(figure 4.3.1.3.5), moreover, it was also observed a trend for an increase in 5-HT turnover

rate [Z=-1.922 (2-tailed) p=0.055] (Table 4.3.1.3.5). In saline male group there was an

increase in the 5-HT turnover rate from 5 to 10 days after the last administration [Z=-2.286

(2-tailed) p<0.05]. Ten days after withdrawal, cocaine-treated males displayed lower 5-HT

turnover rate as compared to cocaine-treated females [Z=-2.082 (2-tailed) p<0.05]. In the

saline group this difference did not reach significance.

Hippocampal levels of HVA were not detectable by HPLC-EC in a considerable

number of samples. Therefore, in this brain region, only the levels of DA metabolite

DOPAC were considered in the analysis.

a a

205

Prefrontal cortex

A

0

10

20

30

40

DA DOPAC HVA DA DOPAC HVA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

5 days 10 days

B

0

50

100

150

200

250

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

5 days 10 days

Figure 4.3.1.3.4- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the prefrontal cortex,

determined by HPLC-EC for male and female rats, 5 and 10 days after withdrawal. Each column represents

the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for at least 5 animals

per group.

206

Hippocampus

A

0

5

10

15

20

DA DOPAC DA DOPAC

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

5 days 10 days

B

0

100

200

300

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

5 days 10 days

Figure 4.3.1.3.5- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and on (B) the levels of 5-HT and its metabolite 5-HIAA in the hippocampus determined

by HPLC-EC for male and female rats, 5 and 10 days after withdrawal. Each column represents the mean +

S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for at least 6 animals per group.

Columns with the same letter are significantly different from each other. a,b p<0.05.

a

a

b b

207

Table 4.3.1.3.4- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the prefrontal cortex, 5 and 10 days after the end of cocaine exposure period.

5 days 10 days

Male Female Male Female

saline cocaine saline cocaine saline cocaine saline cocaine

DOPAC/DA 0.31±0.11

(n=7)

0.40±0.15

(n=8)

0.41±0.13

(n=8)

0.56±0.15

(n=7)

0.38±0.09

(n=5)

0.31±0.03

(n=6)

0.31±0.03

(n=6)

0.36±0.05

(n=7)

HVA/DA 0.54±0.06

(n=7)

0.54±0.07

(n=8)

0.58±0.08

(n=8)

0.57±0.12

(n=7)

0.47±0.15

(n=5)

0.49±0.09

(n=6)

0.58±0.11

(n=6)

0.43±0.07

(n=7)

(DOPAC+HVA)/DA 0.85±0.17

(n=7)

0.95±0.17

(n=8)

0.99±0.14

(n=8)

1.13±0.22

(n=7)

0.85±0.24

(n=5)

0.80±0.09

(n=6)

0.88±0.12

(n=6)

0.79±0.09

(n=7)

5-HIAA/5-HT 0.09±0.02

(n=7)

0.12±0.03

(n=8)

0.09±0.02

(n=8)

0.14±0.08

(n=7)

0.08±0.02

(n=5)

0.11±0.03

(n=6)

0.21±0.08

(n=6)

0.22±0.08

(n=7)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid.

Table 4.3.1.3.5- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA] and [5-HIAA]/[5-HT], in the

hippocampus, 5 and 10 days after the end of cocaine exposure period.

5 days 10 days

Male Female Male Female

saline cocaine saline cocaine saline cocaine saline cocaine

DOPAC/DA 0.73±0.38

(n=7)

0.70±0.29

(n=9)

0.84±0.23

(n=7)

0.91±0.42

(n=6)

0.24±0.05

(n=6)

0.41±0.11

(n=6)

0.54±0.16

(n=6)

0.45±0.13

(n=6)

5-HIAA/5-HT 0.18±0.05

(n=7)

0.24±0.04

(n=9)

0.21±0.02

(n=7)

0.21±0.04

(n=6)

0.41±0.07

(n=6)

0.23±0.04

(n=6)

0.24±0.04

(n=6)

0.48±0.09

(n=6)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. In the same line, values with

same letter are significantly different. DA- dopamine; DOPAC- 3,4-dihydroxyphenylacetic acid; 5-HT-

serotonin; 5-HIAA- 5-hydroxyindolacetic acid. a,b p<0.05.

Within the amygdala, 10 days after withdrawal, cocaine-treated females displayed

lower levels of DOPAC as compared to control females [Z=-2.191 (2-tailed) p<0.05], and

saline males presented higher levels of DOPAC as compared to saline-treated females

[Z=-2.191 (2-tailed) p<0.05] (figure 4.3.1.3.6). In saline-treated female group the levels of

DA and DOPAC decreased from 5 to 10 days after the last administration ([Z=-2.030 (2-

tailed) p<0.05] and [Z=-2.030 (2-tailed) p<0.05], respectively). No differences were found

in the data from 5-HT system between the experimental groups.

b b a a

208

Amygdala

A

DA DOPAC HVA DA DOPAC HVA

0

10

20

30

80160240320

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

B

0

100

200

300

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

5 days 10 days

Figure 4.3.1.3.6- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the amygdala,

determined by HPLC-EC for male and female rats, 5 and 10 days after withdrawal. Each column represents

the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for at least 5 animals

per group. Columns with the same letter are significantly different from each other. a,b,c,d p<0.05.

Within the hypothalamus, no statistical significant differences were found on the

levels of DA, 5-HT and their metabolites or their turnover between the experimental

groups (figure 4.3.1.3.7 and table 4.3.1.3.7).

c b, c, d

d

a

a b

5 days 10 days

saline male cocaine male saline female cocaine female

209

Hypothalamus

A

0

100

200

300

400

DA DOPAC HVA DA DOPAC HVA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

5 days 10 days

B

0

200

400

600

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

5 days 10 days

Figure 4.3.1.3.7- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the hypothalamus,

determined by HPLC-EC for male and female rats, 5 and 10 days after withdrawal. Each column represents

the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for at least 5 animals

per group.

210

Table 4.3.1.3.6- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the amygdala, 5 and 10 days after the end of cocaine exposure period.

5 days 10 days

Male Female Male Female

saline cocaine saline cocaine saline cocaine saline cocaine

DOPAC/DA 0.33±0.15

(n=7)

0.25±0.06

(n=9)

0.34±0.12

(n=7)

0.30±0.10

(n=6)

0.33±0.19

(n=6)

0.34±0.13

(n=6)

0.25±0.11

(n=5)

0.44±0.27

(n=6)

HVA/DA 0.14±0.05

(n=7)

0.17±0.08

(n=9)

0.12±0.03

(n=7)

0.18±0.14

(n=6)

0.08±0.04

(n=6)

0.08±0.02

(n=6)

0.08±0.02

(n=5)

0.07±0.02

(n=6)

(DOPAC+HVA)/DA 0.47±0.14

(n=7)

0.42±0.10

(n=9)

0.47±0.14

(n=7)

0.49±0.16

(n=6)

0.41±0.19

(n=6)

0.42±0.13

(n=6)

0.34±0.11

(n=5)

0.51±0.28

(n=6)

5-HIAA/5-HT 0.30±0.11

(n=7)

0.39±0.16

(n=9)

0.28±0.07

(n=7)

0.29±0.08

(n=6)

0.30±0.09

(n=6)

0.47±0.15

(n=6)

0.31±0.07

(n=5)

0.36±0.10

(n=6)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid.

Table 4.3.1.3.7- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the hypothalamus, 5 and 10 days after the end of cocaine exposure period.

5 days 10 days

Male Female Male Female

saline cocaine saline cocaine saline cocaine saline cocaine

DOPAC/DA 0.26±0.07

(n=8)

0.27±0.07

(n=9)

0.34±0.10

(n=7)

0.37±0.06

(n=6)

0.43±0.12

(n=6)

0.41±0.09

(n=5)

0.41±0.08

(n=5)

0.48±0.13

(n=7)

HVA/DA 0.09±0.04

(n=8)

0.06±0.02

(n=9)

0.09±0.03

(n=7)

0.14±0.11

(n=6)

0.03±0.01

(n=6)

0.07±0.03

(n=5)

0.03±0.01

(n=5)

0.12±0.06

(n=7)

(DOPAC+HVA)/DA 0.35±0.07

(n=8)

0.34±0.08

(n=9)

0.43±0.10

(n=7)

0.50±0.12

(n=6)

0.45±0.12

(n=6)

0.48±0.11

(n=5)

0.44±0.08

(n=5)

0.60±0.18

(n=7)

5-HIAA/5-HT 0.25±0.03

(n=8)

0.30±0.05

(n=9)

0.28±0.08

(n=7)

0.43±0.12

(n=6)

0.31±0.09

(n=6)

0.37±0.10

(n=5)

0.25±0.03

(n=5)

0.25±0.03

(n=7)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid.

Within the dorsal raphe nucleus, statistical analysis revealed that 5 days after the

end of exposure period saline-treated males displayed higher levels of DA as compared to

saline-treated females [Z=-2.662 (2-tailed) p<0.01] (figure 3.3.1.3.8). No other differences

were found between the experimental groups in this brain area.

211

Dorsal raphe nucleus

A

0

100

200

300

DA DOPAC HVA DA DOPAC HVA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

5 days 10 days

B

0

350

700

1050

1400

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

5 days 10 days

Figure 4.3.1.3.8- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the dorsal raphe

nucleus, determined by HPLC-EC for male and female rats, 5 and 10 days after withdrawal. Each column

represents the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for at least

6 animals per group. Columns with the same letter are significantly different from each other. a p<0.01.

a

a

212

Table 4.3.1.3.8- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the dorsal raphe nucleus, 5 and 10 days after the end of cocaine exposure period.

5 days 10 days

Male Female Male Female

saline cocaine saline cocaine saline cocaine saline cocaine

DOPAC/DA 0.50±0.24

(n=8)

0.50±0.16

(n=9)

0.46±0.12

(n=7)

0.56±0.32

(n=7)

0.89±0.39

(n=6)

0.57±0.24

(n=6)

0.73±0.31

(n=6)

0.61±0.23

(n=7)

HVA/DA 0.18±0.07

(n=8)

0.20±0.08

(n=9)

0.16±0.12

(n=7)

0.10±0.05

(n=7)

0.10±0.06

(n=6)

0.12±0.04

(n=6)

0.11±0.03

(n=6)

0.05±0.02

(n=7)

(DOPAC+HVA)/DA 0.69±0.24

(n=8)

0.70±0.15

(n=9)

0.63±0.15

(n=7)

0.66±0.37

(n=7)

0.99±0.41

(n=6)

0.70±0.27

(n=6)

0.84±0.32

(n=6)

0.66±0.25

(n=7)

5-HIAA/5-HT 0.32±0.06

(n=8)

0.29±0.05

(n=9)

0.46±0.21

(n=7)

0.31±0.07

(n=7)

0.67±0.37

(n=6)

0.87±0.59

(n=6)

0.28±0.05

(n=6)

0.37±0.05

(n=7)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid.

Within the cerebellum, statistical analysis revealed that in the saline male group

the levels of HVA decreased from 5 to 10 days after the last administration [Z=-2.143 (2-

tailed) p<0.05] (figure 4.3.1.3.9). No other differences were found between the

experimental groups in the monoamine levels.

213

Cerebellum

A

0

10

20

30

DA DOPAC HVA DA DOPAC HVA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

5 days 10 days

B

0

20

40

60

80

100

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male saline female cocaine female

5 days 10 days

Figure 4.3.1.3.9- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the cerebellum,

determined by HPLC-EC for male and female rats, 5 and 10 days after withdrawal. Each column represents

the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for at least 6 animals

per group. Columns with the same letter are significantly different from each other. a p<0.05.

a

a

214

Table 4.3.1.3.9- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the cerebellum, 5 and 10 days after the end of cocaine exposure period.

5 days 10 days

Male Female Male Female

saline cocaine saline cocaine saline cocaine saline cocaine

DOPAC/DA 0.74±0.22

(n=7)

1.12±0.42

(n=9)

0.67±0.18

(n=7)

1.71±0.53

(n=8)

1.21±0.28

(n=6)

1.71±1.19

(n=6)

1.84±1.28

(n=6)

0.74±0.15

(n=6)

HVA/DA 1.53±0.80

(n=7)

0.92±0.12

(n=9)

0.60±0.18

(n=7)

0.55±0.13

(n=8)

1.24±0.35

(n=6)

0.68±0.21

(n=6)

0.91±0.23

(n=6)

1.34±0.62

(n=6)

(DOPAC+HVA)/DA 2.29±0.77

(n=7)

2.03±0.37

(n=9)

1.27±0.26

(n=7)

2.26±0.44

(n=8)

2.45±0.58

(n=6)

2.39±1.37

(n=6)

2.75±1.42

(n=6)

2.08±0.75

(n=6)

5-HIAA/5-HT 0.52±0.14

(n=7)

0.25±0.03

(n=9)

0.22±0.04

(n=7)

0.36±0.10

(n=8)

0.30±0.06

(n=6)

0.22±0.06

(n=6)

0.23±0.04

(n=6)

0.34±0.08

(n=6)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid.

Overall, our basal neurochemical and hormonal data point to much more pronounced

long-term effects of cocaine in male rats than in females. Thus, further testing, namely

determination of dopaminergic-related, HPA-related and HPG-related mRNA expression

levels and behavioural studies, such as evaluation of anxiety-like and aggressive

behaviour were only performed in male rats.

4.3.1.4. mRNA expression levels determination

4.3.1.4.1. Dopaminergic-related mRNA expression levels

The expression levels of mRNAs related with synthesis (TH), metabolization

(MAO-A), transportation (DAT) of DA, and D1, D2 and D3 receptors mRNA were

assessed by qRT-PCR in male rats at 10 days after the end of exposure protocol.

Data analysis revealed that the expression levels of TH mRNA were increased in

cocaine exposed males in the SN-VTA as compared to control males [t(4.372)=-3.089;

p<0.04] (figure 4.3.1.4.1.1.). In this brain area no differences were observed in the

expression levels of the other mRNA analysed, although a trend towards increased DAT

mRNA expression was observed in cocaine-treated animals [t(10)=-2.064; p=0.066]. In

the other brain areas studied, Nac, dorsal striatum, prefrontal cortex hippocampus,

amygdala and hypothalamus, the levels of mRNA expression of TH, MAO-A, DAT and DA

receptors D1, D2 and D3 did not differ between the cocaine-treated animals and control

animals (from figure 4.3.1.4.1.1. to figure 4.3.1.4.1.7.)

215

SN-VTA

A B

0.0000.0030.0060.0090.012

TH

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

0.000

0.002

0.004

0.006

DAT

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

C D

0.0000.0020.0040.0060.008

MAO-A

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

0.00000

0.00003

0.00006

0.00009

D1

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

E F

0.0000

0.0002

0.0004

0.0006

D2

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

0.0000000.0000050.0000100.0000150.000020

D3

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

Figure 4.3.1.4.1.1. Effects of chronic cocaine exposure throughout adolescence on the levels of mRNA

expression of (A) TH, (B) DAT, (C) MAO-A, (D) D1, (E) D2 and (F) D3 in the SN-VTA, determined by qRT-

PCR for male rats 10 days after withdrawal. Each column represents the mean + S.E.M. of normalized mRNA

expression, for at least 5 animals per group. Columns with the same letter are significantly different from each

other. a p<0.05.

a

a

216

Nucleus accumbens

A B

0.00000

0.00002

0.00004

0.00006

TH

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

0.00000

0.00001

0.00002

0.00003

0.00004

DAT

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

C D

0.00

0.01

0.02

0.03

0.04

MAO-A

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

0.0

0.3

0.6

0.9

1.2

D1

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

E F

0.00

0.01

0.02

0.03

D2

norm

aliz

ed m

RN

A

expr

esso

n le

vels

saline male cocaine male

0.00000.00020.00040.00060.00080.0010

D3

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

Figure 4.3.1.4.1.2. Effects of chronic cocaine exposure throughout adolescence on the levels of mRNA

expression of (A) TH, (B) DAT, (C) MAO-A, (D) D1, (E) D2 and (F) D3 in the Nac, determined by qRT-PCR for

male rats 10 days after withdrawal. Each column represents the mean + S.E.M. of normalized mRNA

expression, for at least 5 animals per group.

217

Dorsal striatum

A B

0.000000

0.000006

0.000012

0.000018

TH

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

0.000000

0.000002

0.000004

0.000006

0.000008

DAT

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

C D

0.000

0.025

0.050

0.075

MAO-A

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

0.000

0.025

0.050

0.075

D1

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

E F

0.0000.0050.0100.0150.0200.025

D2

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

0.0000

0.0001

0.0002

0.0003

0.0004

D3

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

Figure 4.3.1.4.1.3. Effects of chronic cocaine exposure throughout adolescence on the levels of mRNA

expression of (A) TH, (B) DAT, (C) MAO-A, (D) D1, (E) D2 and (F) D3 in the dorsal striatum, determined by

qRT-PCR for male rats 10 days after withdrawal. Each column represents the mean + S.E.M. of normalized

mRNA expression, for at least 5 animals per group.

218

Prefrontal cortex

A B

0.0000

0.0001

0.0002

0.0003

DAT

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

0.0000.0050.0100.0150.0200.025

MAO-A

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

C D

0.00000.00020.00040.00060.00080.0010

D1

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

0.00000.00030.00060.00090.00120.0015

D2

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

E

0.00000

0.00006

0.00012

0.00018

D3

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

Figure 4.3.1.4.1.4. Effects of chronic cocaine exposure throughout adolescence on the levels of mRNA

expression of (A) DAT, (B) MAO-A, (C) D1, (D) D2 and (E) D3 in the prefrontal cortex, determined by qRT-

PCR for male rats 10 days after withdrawal. Each column represents the mean + S.E.M. of normalized mRNA

expression, for at least 5 animals per group.

219

Hippocampus

A B

0.00000

0.00001

0.00002

0.00003

DAT

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

0.000.020.040.060.080.10

MAO-A

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

C D

0.000

0.005

0.010

0.015

0.020

D1

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

0.0000.0010.0020.0030.004

D2

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

E

0.000000.000020.000040.000060.000080.00010

D3

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

Figure 4.3.1.4.1.5. Effects of chronic cocaine exposure throughout adolescence on the levels of mRNA

expression of (A) DAT, (B) MAO-A, (C) D1, (D) D2 and (E) D3 in the hippocampus, determined by qRT-PCR

for male rats 10 days after withdrawal. Each column represents the mean + S.E.M. of normalized mRNA

expression, for at least 5 animals per group.

220

Amygdala

A B

0.0000000

0.0000002

0.0000004

0.0000006

DAT

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

0.000

0.003

0.006

0.009

MAO-A

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

C D

0.0000

0.0002

0.0004

0.0006

D1

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

0.00000

0.00004

0.00008

0.00012

0.00016

D2

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

E

0.00000000.00000030.00000060.00000090.0000012

D3

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

Figure 4.3.1.4.1.6. Effects of chronic cocaine exposure throughout adolescence on the levels of mRNA

expression of (A) DAT, (B) MAO-A, (C) D1, (D) D2 and (E) D3 in the amygdala, determined by qRT-PCR for

male rats 10 days after withdrawal. Each column represents the mean + S.E.M. of normalized mRNA

expression, for at least 5 animals per group.

221

Hypothalamus

A B

0.00000.00020.00040.00060.00080.0010

DAT

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

0.00

0.06

0.12

0.18

MAO-A

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

C D

0.00

0.01

0.02

0.03

0.04

D1

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

0.000

0.001

0.002

0.003

0.004

D2

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

E

0.0000

0.0001

0.0002

0.0003

D3

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

Figure 4.3.1.4.1.7. Effects of chronic cocaine exposure throughout adolescence on the levels of mRNA

expression of (A) DAT, (B) MAO-A, (C) D1, (D) D2 and (E) D3 in the hypothalamus, determined by qRT-PCR

for male rats 10 days after withdrawal. Each column represents the mean + S.E.M. of normalized mRNA

expression, for at least 5 animals per group.

4.3.1.4.2. HPA and HPG axes-related mRNA expression levels

The expression levels of mRNAs related with HPA and HPG axes were assessed

by qRT-PCR in male rats 10 days after the end of the exposure protocol.

Data analysis revealed that the expression levels of GR mRNA did not differ

between cocaine-treated males and control males in all the brain areas studied, excepted

222

for the SN-VTA, where cocaine-treated males presented lower levels of GR mRNA as

compared to control males [t(8.353)=2,329; p<0.05] (figure 4.3.1.4.2.1).

Within the hypothalamus, statistical analysis by Student’s t-test showed no

differences in the mRNA expression levels of CRH and AR in the cocaine-treated males

as compared to control males (figure 4.3.1.4.2.2).

Glucocorticoids receptor

sn-vta Nac DStriatum PFC Hippoc. Amygd. Hip. Pit.

0.000

0.002

0.004

0.006

0.04

0.08

norm

aliz

ed m

RN

A e

xpre

ssio

n le

vels

Figure 4.3.1.4.2.1. Effects of chronic cocaine exposure throughout adolescence on the levels of mRNA

expression of GR in the SN-VTA, Nac, dorsal striatum, prefrontal cortex, hippocampus, amygdala,

hypothalamus and pituitary, determined by qRT-PCR for male rats 10 days after withdrawal. Each column

represents the mean + S.E.M. of normalized mRNA expression, for at least 5 animals per group. a p<0.05.

Hypothalamus

A B

0.00

0.01

0.02

0.03

0.04

CRH

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

0.00

0.01

0.02

0.03

AR

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

Figure 4.3.1.4.2.2. Effects of chronic cocaine exposure throughout adolescence on the levels of mRNA

expression of (A) CRH and (B) AR, in the hypothalamus, determined by qRT-PCR for male rats 10 days after

withdrawal. Each column represents the mean + S.E.M. of normalized mRNA expression, for at least 5

animals per group.

saline male cocaine male

a a

223

Within the pituitary, the mRNA expression levels of CRH-R1, GnRH-R, AR, POMC

and LHβ were studied and data analysis did not reveal differences between cocaine- and

saline-treated male rats in none of mRNAs studied (figure 4.3.1.4.2.3).

Pituitary

A B

0.0000.0040.0080.0120.016

CRH-R1

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

0.00

0.02

0.04

0.06

GnRH-R

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

C D

0.0000.0050.0100.0150.020

AR

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

020406080

100

POMC

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

E

02468

LH

norm

aliz

ed m

RN

A

expr

essi

on le

vels

saline male cocaine male

Figure 4.3.1.4.2.3. Effects of chronic cocaine exposure throughout adolescence on the levels of mRNA

expression of (A) CRH-R1, (B) GnRH-R, (C) AR, (D) POMC and (E) LH in the pituitary, determined by qRT-

PCR for male rats 10 days after withdrawal. Each column represents the mean + S.E.M. of normalized mRNA

expression, for at least 5 animals per group.

224

4.3.2. Behavioural studies and associated measures

4.3.2.1. Anxiety-like behaviour

Elevated plus maze test. Five and 10 days after cocaine withdrawal male rats were

tested in the EPM apparatus, in a 5 minutes session. EPM behavioural data was analysed

by a 2-way ANOVA (treatment x time point of assessment).

Statistical analysis revealed that cocaine-treated males presented no differences in

frequency of open and closed arm entries and in frequency of central platform entries,

neither 5 or 10 days after withdrawal (figure 4.3.2.1.1). Also no differences were observed

between cocaine and saline-treated male rats on the time spent in the open and closed

arms, neither 5 or 10 days after withdrawal (figure 4.3.2.1.2). ANOVA revealed a main

effect of treatment in the time spent in the central platform [F(1,32)=6.326; p<0.02].

Cocaine-treated male rats spent less time as compared to controls. Statistical analysis

also revealed that 10 days after withdrawal cocaine-treated male rats spent less time in

the central platform than controls [t(19.827)=-3.124; p<0.01] (figure 4.3.2.1.3 B).

A B

02468

5 days 10 days

Num

ber o

f ent

ries

saline male cocaine male

0

20

40

60

5 days 10 days

% o

f ent

ries

saline male cocaine male

C D

0

3

6

9

5 days 10 days

Num

ber o

f ent

ries

saline male cocaine male

048

1216

5 days 10 days

Num

ber o

f ent

ries

saline male cocaine male

Figure 4.3.2.1.1- Effects of chronic cocaine exposure throughout adolescence on the number of arm entries

during the elevated plus maze test performed 5 and 10 days after withdrawal by male rats. A- Number of open

arm entries; B- percentage of open arm entries; C- Number of closed arm entries; D- Number of total arm

entries. Each column represents the mean + S.E.M., for at least 7 animals per group.

Open arms Open arms

Closed arms Total arm entries

225

A B

0306090

120150

5 days 10 days

Dur

atio

n (s

ec.)

saline male cocaine male

050

100150200250

5 days 10 days

Dur

atio

n (s

ec.)

saline male cocaine male

Figure 4.3.2.1.2- Effects of chronic cocaine exposure throughout adolescence on the time spent (A) in open

arms and (B) in closed arms during the elevated plus maze test performed 5 and 10 days after withdrawal, by

male rats. Each column represents the mean + S.E.M., for at least 7 animals per group.

A B

048

1216

5 days 10 days

Num

ber o

f ent

ries

saline male cocaine male

0

20

40

60

5 days 10 days

Dur

atio

n (s

ec.)

saline male cocaine male

Figure 4.3.2.1.3- Effects of chronic cocaine exposure throughout adolescence (A) in the number of central

platform entries and (B) in the time spent in the central platform during the elevated plus maze test performed

5 and 10 days after cocaine withdrawal, by male rats. Each column represents the mean + S.E.M., for at least

7 animals per group. Columns with the same letter are significantly different from each other. a p<0.01.

No group differences were observed in frequency and duration of risk-like

behaviour, rearing, head-dipping and grooming during the EPM (table 4.3.2.1.1).

Open arms Closed arms

Central platform Central platform a

a

226

Table 4.3.2.1.1- Frequency and duration of behavioural categories like risk-like behaviour, rearing, head

dipping and grooming, during the 5 minutes of the elevated plus maze test, in saline- and cocaine-treated

male rats, 5 and 10 days after the last administration.

5 days 10 days

male saline

(n=7) male cocaine

(n=7) male saline

(n=11) male cocaine

(n=11)

Risk-like Behaviour frequency 0.14±0.14 0.29±0.18 0.09±0.09 0.36±0.15

duration (sec.) - - - -

Rearing frequency 15.14±1.53 16.00±2.41 16.18±1.50 13.45±1.83

duration (sec.) 32.49±4.89 39.11±6.49 42.74±4.75 31.98±4.89

Head-dipping frequency 1.86±0.67 2.71±0.36 4.73±1.10 3.36±1.05

duration (sec.) 1.30±0.58 2.74±0.71 4.75±1.68 2.68±1.07

Grooming frequency 1.71±0.36 1.71±0.42 1.27±0.19 2.09±0.65

duration (sec.) 12.34±4.21 8.57±2.47 7.90±1.87 11.30±3.70

Data expressed as the mean value ± S.E.M.; n, number of animals per group.

Corticosterone plasma levels. The animals that performed the EPM 10 days after

the end of the exposure protocol were decapitated after the behavioural test and levels of

corticosterone were measured in the obtained plasma. Statistical significance between

groups was assessed using the Student’s t-test. Analysis revealed that, 10 days after

withdrawal, cocaine-treated males had increased corticosterone plasma levels induce

after the EPM (t(16)=2.661; p<0.02) (figure 4.3.2.1.4).

0

100

200

300

Cor

ticos

tero

ne

leve

ls n

g/m

L

saline male cocaine male

Figure 4.3.2.1.4- Effects of chronic cocaine exposure throughout adolescence on the alterations of the

corticosterone plasma levels induced by the elevated plus maze test 10 days after withdrawal, in male rats.

Each column represents the mean + S.E.M for at least 8 animals per group. Columns with the same letter are

significantly different from each other. a p<0.02.

a

a

227

Neurochemical determinations. The levels of DA, 5-HT and their metabolites,

DOPAC, HVA and 5-HIAA were measured by HPLC-EC, in the prefrontal cortex, the

hippocampus, the amygdala, the hypothalamus, the SN-VTA and the dorsal raphe

nucleus after the EPM 10 days after the end of exposure protocol. The data obtained after

the test and the basal data obtained 10 days after the end of the exposure protocol were

analyzed by the non-parametric Mann-Whitney U Test.

Within the SN-VTA, no significant differences were found on the levels of DA, 5-

HT, their metabolites or in the turnover rates between cocaine exposed and control males,

after the EPM (figure 4.3.2.1.5. and table 4.3.2.1.2). Also no significant differences were

found between basal levels and levels after the EPM.

Within the dorsal raphe nucleus, no significant differences were found on the levels

of DA, 5-HT, their metabolites or in the turnover rates between cocaine- and saline-treated

male rats after the EPM (figure 4.3.2.1.6. and table 4.3.2.1.3). After the EPM, in this brain

area, the levels of HVA were not detectable by the HPLC-EC. Statistical analysis revealed

that the levels 5-HT were higher after the EPM as compared to basal data, both in saline

and cocaine males ([Z=-2.003 (2-tailed) p<0.05] and [Z=-2.475 (2-tailed) p<0.02],

respectively) (figure 4.3.2.1.6). After the EPM, saline-treated male presented decreased 5-

HIAA/5-HT turnover rate as compared to basal conditions [Z=-2.003 (2-tailed) p<0.05]

(table 4.3.2.1.3).

228

SN-VTA

A

0

200

400

600

800

DA DOPAC HVA DA DOPAC HVA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male

Basal EPM

B

0

150

300

450

600

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rost

rans

mitt

ers

conc

entr

atio

n (n

g/m

g of

pro

tein

)

saline male cocaine male

EPMBasal

Figure 4.3.2.1.5- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the SN-VTA,

determined 10 days after withdrawal, under basal and anxiogenic (EPM) by HPLC-EC for male rats. Each

column represents the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for

at least 6 animals per group. Columns with the same letter are significantly different from each other. a p<0.05.

a a

229

Dorsal raphe nucleus

A

0

100

200

300

DA DOPAC HVA DA DOPAC HVA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male

Basal EPM

B

0

250

500

750

1000

1250

1500

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male

EPMBasal

Figure 4.3.2.1.6- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the dorsal raphe

nucleus, determined 10 days after withdrawal, under basal and anxiogenic (EPM) by HPLC-EC for male rats.

Each column represents the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of

protein, for at least 6 animals per group. nd- not detectable. Columns with the same letter are significantly

different from each other. a p<0.05, b p<0.02.

b

b

nd

a

a

230

Table 4.3.2.1.2- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the SN-VTA in saline- and cocaine-treated male rats, as measured 10 days after the last

administration, under basal condition and after the elevated plus maze test.

Basal EPM

saline male cocaine male saline male cocaine male

DOPAC/DA 0.24±0.06

(n=6)

0.16±0.02

(n=6)

0.21±0.01

(n=11)

0.20±0.01

(n=9)

HVA/DA 0.09±0.02

(n=6)

0.07±0.02

(n=6)

0.12±0.01

(n=11)

0.09±0.02

(n=9)

(DOPAC+HVA)/DA 0.33±0.05

(n=6)

0.24±0.04

(n=6)

0.33±0.02

(n=11)

0.28±0.03

(n=9)

5-HIAA/5-HT 6.55±6.27

(n=6)

0.22±0.03

(n=6)

0.23±0.02

(n=11)

0.27±0.03

(n=9)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid.

Table 4.3.2.1.3- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the dorsal raphe nucleus in saline- and cocaine-treated male, as measured 10 days after

the last administration, under basal condition and after the elevated plus maze test.

Basal EPM

saline male cocaine male saline male cocaine male

DOPAC/DA 0.89±0.39

(n=6)

0.57±0.24

(n=6)

0.31±0.02

(n=9)

0.25±0.03

(n=9)

HVA/DA 0.10±0.06

(n=6)

0.12±0.04

(n=6) - -

(DOPAC+HVA)/DA 0.99±0.41

(n=6)

0.70±0.27

(n=6)

- -

5-HIAA/5-HT 0.67±0.37

(n=6)

0.87±0.59

(n=6)

0.24±0.01

(n=9)

0.24±0.02

(n=9)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid. In

the same line, values with same letter are significantly different. a p<0.05.

Within the prefrontal cortex, no significant differences were found between

cocaine- and saline-treated male rats after the EPM (figure 4.3.2.1.7. and table 4.3.2.1.4).

Statistical analysis revealed that both saline- and cocaine-treated males after the EPM

presented higher HVA/DA and (DOPAC+HVA)/DA turnover rates as compared to animals

under basal conditions (saline males- HVA/DA [Z=-2.209 (2-tailed) p<0.05],

(DOPAC+HVA)/DA [Z=-2.209 (2-tailed) p<0.05]; cocaine males- HVA/DA [Z=-2.711 (2-

tailed) p<0.01], (DOPAC+HVA)/DA [Z=-2.828 (2-tailed) p<0.01]). However, cocaine-

a a

231

treated males after the EPM presented higher DOPAC/DA turnover rate as compared to

cocaine male rats under basal conditions [Z=-2.593 (2-tailed) p<0.02] and this effect was

not observed in saline-treated male rats.

Prefrontal cortex

A

0

10

20

30

40

DA DOPAC HVA DA DOPAC HVA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male

Basal EPM

B

0

100

200

300

400

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male

EPMBasal

Figure 4.3.2.1.7- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the prefrontal cortex,

determined 10 days after withdrawal, under basal and anxiogenic (EPM) by HPLC-EC for male rats. Each

column represents the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for

at least 5 animals per group.

232

Table 4.3.2.1.4- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the prefrontal cortex in saline- and cocaine-treated male, as measured 10 days after the

last administration, under basal condition and after the elevated plus maze test.

Basal EPM

saline male cocaine male saline male cocaine male

DOPAC/DA 0.38±0.09

(n=5)

0.31±0.03

(n=6)

0.68±0.13

(n=11)

2.65±1.18

(n=9)

HVA/DA 0.47±0.15

(n=5)

0.49±0.09

(n=6)

0.89±0.08

(n=11)

3.58±2.59

(n=9)

(DOPAC+HVA)/DA 0.85±0.24

(n=5)

0.80±0.09

(n=6)

1.57±0.10

(n=11)

5.83±3.24

(n=9)

5-HIAA/5-HT 0.08±0.02

(n=5)

0.11±0.03

(n=6)

0.08±0.01

(n=11)

0.11±0.01

(n=9)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid. In

the same line, values with same letter are significantly different. a p<0.02, b,d p<0.05, c,e p<0.01.

Within the hippocampus, no significant differences were found on the levels of DA,

5-HT, their metabolites and in turnover rates between cocaine- and saline-treated male

rats after the EPM (figure 4.3.2.1.8. and table 4.3.2.1.5). However, the saline-treated

males after EPM presented lower 5-HIAA/5-HT turnover rate as compared to saline males

under basal conditions [Z=-2.234 (2-tailed) p<0.05], moreover, in the cocaine-treated

animals this difference did not reach significance.

Within the amygdala, after the EPM, cocaine-treated males presented lower levels

of DOPAC and 5-HIAA as compared to saline-treated males ([Z=-3.009 (2-tailed) p<0.005]

and [Z=-1.967 (2-tailed) p<0.05], respectively) (figure 4.3.2.1.9). Saline-treated males

presented increased levels of DOPAC after EPM, as compared to data from basal

conditions [Z=-2.711 (2-tailed) p<0.01], this effects was not observed in cocaine-treated

animals.

a a

c c

e e

b b

d d

233

Hippocampus

A

0

5

10

15

20

DA DOPAC DA DOPAC

Neu

rost

rans

mitt

ers

conc

entr

atio

n (n

g/m

g of

pro

tein

)

saline male cocaine male

Basal EPM

B

0

100

200

300

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s C

once

ntra

tion

(ng/

mg

of p

rote

in)

saline male cocaine male

EPMBasal

Figure 4.3.2.1.8 - Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolite DOPAC and on (B) the levels of 5-HT and its metabolite 5-HIAA in the hippocampus, determined

10 days after withdrawal, under basal and anxiogenic (EPM) by HPLC-EC for male rats. Each column

represents the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for at least

6 animals per group.

234

Amygdala

A

0

40

80

120

DA DOPAC HVA DA DOPAC HVA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male

Basal EPM

B

0

75

150

225

300

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male

EPMBasal

Figure 4.3.2.1.9- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the amygdala,

determined 10 days after withdrawal, under basal and anxiogenic (EPM) by HPLC-EC for male rats. Each

column represents the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of protein, for

at least 6 animals per group. Columns with the same letter are significantly different from each other. a p<0.01, b p<0.005, c p<0.05.

a,b

b

c c

a

235

Table 4.3.2.1.5- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA] and [5-HIAA]/[5-HT], in the

hippocampus in saline- and cocaine-treated male, as measured 10 days after the last administration, under

basal condition and after the elevated plus maze test.

Basal EPM

saline male cocaine male saline male cocaine male

DOPAC/DA 0.24±0.05

(n=6)

0.41±0.11

(n=6)

0.39±0.06

(n=8)

0.42±0.10

(n=6)

5-HIAA/5-HT 0.41±0.07

(n=6)

0.23±0.04

(n=6)

0.18±0.03

(n=8)

0.16±0.03

(n=6)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid. In the same line, values with

same letter are significantly different. a p<0.05.

Table 4.3.2.1.6- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the amygdala in saline- and cocaine-treated male, as measured 10 days after the last

administration, under basal condition and after the elevated plus maze test.

Basal EPM

saline male cocaine male saline male cocaine male

DOPAC/DA 0.33±0.19

(n=6)

0.34±0.13

(n=6)

0.37±0.04

(n=8)

0.36±0.07

(n=7)

HVA/DA 0.08±0.04

(n=6)

0.08±0.02

(n=6)

0.11±0.04

(n=8)

0.15±0.06

(n=7)

(DOPAC+HVA)/DA 0.41±0.19

(n=6)

0.42±0.13

(n=6)

0.49±0.04

(n=8)

0.51±0.10

(n=7)

5-HIAA/5-HT 0.30±0.09

(n=6)

0.47±0.15

(n=6)

0.33±0.07

(n=8)

0.37±0.05

(n=7)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid.

Within the hypothalamus, no significant differences were found on the levels of DA,

5-HT, their metabolites and in turnover rates between cocaine- and saline-treated male

rats after the EPM (figure 4.3.2.1.10. and table 4.3.2.1.7). After the EPM, in this brain area

the levels of HVA were not detectable by the HPLC-EC.

a a

236

Hypothalamus

A

0

50

100

150

200

250

DA DOPAC HVA DA DOPAC HVA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male

Basal EPM

B

0

200

400

600

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ions

(n

g/m

g of

pro

tein

)

saline male cocaine male

EPMBasal

Figure 4.3.2.1.10- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and

its metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the

hypothalamus, determined 10 days after withdrawal, under basal and anxiogenic (EPM) by HPLC-EC for male

rats. Each column represents the mean + S.E.M. of monoamine levels, expressed as ng of monoamine/mg of

protein, for at least 5 animals per group. nd- not detectable. Columns with the same letter are significantly

different from each other. a p<0.05

nd

237

Table 4.3.2.1.7- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the hypothalamus in saline- and cocaine-treated male, as measured 10 days after the last

administration, under basal condition and after the elevated plus maze test.

Basal EPM

saline male cocaine male saline male cocaine male

DOPAC/DA 0.43±0.12

(n=6)

0.41±0.09

(n=5)

0.24±0.02

(n=11)

0.21±0.04

(n=9)

HVA/DA 0.03±0.01

(n=6)

0.07±0.03

(n=5) - -

(DOPAC+HVA)/DA 0.45±0.12

(n=6)

0.48±0.11

(n=5) - -

5-HIAA/5-HT 0.31±0.09

(n=6)

0.37±0.10

(n=5)

0.18±0.02

(n=11)

0.20±0.02

(n=9)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid.

4.3.2.2. Aggressive behaviour

Resident-intruder paradigm. Ten days after cocaine withdrawal, aggressive

behaviour of male rats was evaluated using the resident-intruder paradigm. Behavioural

activity during test was analysed by the Student’s t-test.

Data analysis revealed that cocaine-treated males presented no differences in

frequency, duration and latency of the offensive behaviours (attack, bite, pinning,

wrestling, chasing and aggressive grooming) (figure 4.3.2.2.1). Cocaine males displayed

lower frequency of flight behaviour, a behaviour from the social investigation behaviour

category, [t(14)=-2.430; p<0.05], the latency to this behaviour was also increased in

cocaine-treated males as compared to control males [t(7.493)=2.346; p<0.05] (figure

4.3.2.2.2-A and C). Student’s t-test also revealed that the duration of the anogenital

sniffing behaviour, a behaviour from defensive behaviour category, was increased in

cocaine-treated males [t(14)=2.474; p<0.05] (figure 4.3.2.2.3-B).

238

Offensive behaviour

0

10

20

30

40

50

attack bite pinning w restling chasing aggressivegrooming

Freq

uenc

y

saline male cocaine male

0

30

60

90

120

pinning w restling chasing agressive grooming

Dur

atio

n (s

ec.)

saline male cocaine male

0

200

400

600

attack bite pinning w restling chasing aggressivegrooming

Late

ncy

(sec

.)

saline male cocaine male

Figure 4.3.2.2.1- Effects of chronic cocaine exposure throughout adolescence on (A) frequency, (B) duration

and (C) latency of offensive behaviours exhibited by the resident animal during the resident-intruder paradigm

performed 10 days after the end of exposure protocol. Each column represents the mean + S.E.M., for at least

7 animals per group.

A

B

C

239

Defensive behaviour

0

30

60

90

flight avoidance of contact supine posture

Freq

uenc

y

saline male cocaine male

0

20

40

60

80

100

flight avoidance of contact supine posture

Dur

atio

n (s

ec.)

saline male cocaine male

0

150

300

450

flight avoidance of contact supine posture

Late

ncy

(sec

.)

saline male cocaine male

Figure 4.3.2.2.2- Effects of chronic cocaine exposure throughout adolescence on (A) frequency, (B) duration

and (C) latency of defensive behaviours performed by the resident animal during the resident-intruder

paradigm 10 days after the end of exposure protocol. Each column represents the mean + S.E.M., for at least

7 animals per group. a,b p<0.05

a a

b

b

A

B

C

240

Social investigation behaviour

0

20

40

60

80

100

sniff ing anogenital sniff ing approach behaviour

Freq

uenc

y

saline male cocaine male

0

30

60

90

120

sniff ing anogenital sniff ing approach behaviour

Dur

atio

n (s

ec.)

saline male cocaine male

0

150

300

450

sniff ing anogenital sniff ing approach behaviour

Late

ncy

(sec

.)

saline male cocaine male

Figure 4.3.2.2.3- Effects of chronic cocaine exposure throughout adolescence on (A) frequency, (B) duration

and (C) latency of defensive behaviours performed by the resident animal during the resident-intruder

paradigm 10 days after the end of exposure protocol. Each column represents the mean + S.E.M., for at least

7 animals per group. a p<0.05

a a

A

B

C

241

Corticosterone plasma levels. The animals that performed the resident-intruder

paradigm 10 days after the end of the exposure protocol were decapitated and

corticosterone levels were measured in the obtained plasma. Statistical significance

between groups was assessed using the Student’s t-test. Statistical analysis revealed that

10 days after the end of exposure protocol, the increase in levels of corticosterone

induced by the resident-intruder paradigm did not differ between cocaine- and saline-

treated males (figure 4.3.2.2.4). Importantly, this levels were much higher than the basal.

0

150

300

450

Cor

ticos

tero

ne le

vels

(n

g/m

L)

saline male cocaine male

Figure 4.3.2.2.4- Effects of chronic cocaine exposure throughout adolescence on the alterations on the

plasma levels of corticosterone induced by the resident-intruder paradigm 10 days after withdrawal, in male

rats. Doted line represents the basal levels of corticosterone (see figure 4.3.1.2.1 B). Each column represents

the mean + S.E.M for at least 8 animals per group.

Neurochemical determinations. The levels of DA, 5-HT and their metabolites,

DOPAC, HVA and 5-HIAA were measured by HPLC-EC, in the prefrontal cortex,

hippocampus, amygdala, hypothalamus, SN-VTA and dorsal raphe nucleus after the

behavioural test performed 10 days after the end of exposure protocol. The data obtained

after the test and the basal data 10 days after the end of the exposure protocol were

analyzed by the non-parametric Mann-Whitney U Test.

Within the SN-VTA, after the resident-intruder paradigm cocaine-treated males had

lower 5-HIAA/5-HT turnover rate as compared to saline males [Z=-2.125 (2-tailed) p<0.05]

(table 4.3.2.2.1). Moreover, in the cocaine-treated male group the 5-HIAA/5-HT turnover

rate was decreased after the resident-intruder paradigm as compared to basal data [Z=-

2.125 (2-tailed) p<0.05]. Also in the cocaine-treated male group, the levels of DOPAC and

5-HIAA were decreased after the behavioural test, as compared to basal data ([Z=-2.357

(2-tailed) p<0.02] and [Z=-2.711 (2-tailed) p<0.01], respectively) (figure 4.3.2.2.5). These

differences were not observed in saline-treated males. After the behavioural test, in this

brain area the levels of HVA were not detectable by the HPLC-EC.

242

SN-VTA

A

DA DOPAC HVA DA DOPAC HVA

0

50

100

2000

4000

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

B

0

150

300

450

600

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male

Resident-intruder paradigmBasal

Figure 4.3.2.2.5- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the SN-VTA,

determined 10 days after withdrawal, under basal conditions and after the resident-intruder paradigm, by

HPLC-EC for male rats. Each column represents the mean + S.E.M. of monoamine levels, expressed as ng of

monoamine/mg of protein, for at least 6 animals per group. nd- not detectable. Columns with the same letter

are significantly different from each other. a p<0.05, b p<0.02, c p<0.01.

nd

a

a,b

b

c c

saline male cocaine male

Basal Resident-intruder paradigm

243

Table 4.3.2.2.1- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the SN-VTA in saline- and cocaine-treated male, as measured 10 days after the last

administration, under basal condition and after the resident-intruder paradigm.

Basal Resident-intruder paradigm

saline male cocaine male saline male cocaine male

DOPAC/DA 0.24±0.06

(n=6)

0.16±0.02

(n=6)

0.33±0.15

(n=7)

0.10±0.03

(n=9)

HVA/DA 0.09±0.02

(n=6)

0.07±0.02

(n=6) - -

(DOPAC+HVA)/DA 0.33±0.05

(n=6)

0.24±0.04

(n=6)

- -

5-HIAA/5-HT 6.55±6.27

(n=6)

0.22±0.03

(n=6)

0.22±0.04

(n=7)

0.13±0.01

(n=9)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid. In

the same line, values with same letter are significantly different. a,b p<0.05.

Within the dorsal raphe nucleus, statistical analysis revealed no significant

differences on the levels of DA, 5-HT, their metabolites and in turnover rates between

cocaine exposed and control males after the resident-intruder paradigm (figure 4.3.2.2.6

and table 4.3.2.2.2). Nevertheless, in the saline-treated males the 5-HIAA/5-HT turnover

rate was decreased after the test as compared to basal data [Z=-2.204 (2-tailed) p<0.05].

After the test in this brain area the levels of HVA were not detectable by the HPLC-EC.

Within the prefrontal cortex, no significant differences were found on the levels of

DA, 5-HT, their metabolites and in turnover rates between cocaine exposed and control

males, after the resident-intruder paradigm (figure 4.3.2.2.7 and table 4.3.2.2.3). However,

in cocaine-treated males the levels of HVA were decreased after the behavioural test as

compared to basal data [Z=-2.121 (2-tailed) p<0.05], moreover the HVA/DA and

(DOPAC+HVA)/DA turnover rates were also decreased ([Z=-2.241 (2-tailed) p<0.05] and

[Z=-2.475 (2-tailed) p<0.02], respectively).

a a,b b

244

Dorsal raphe nucleus

A

DA DOPAC HVA DA DOPAC HVA

01020

200

400

600

800N

euro

tran

smitt

ers

conc

entr

atio

n(n

g/m

g of

pro

tein

)

B

0

250

500

750

1000

1250

1500

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male

Resident-intruder paradigmBasal

Figure 4.3.2.2.6- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the dorsal raphe

nucleus, determined 10 days after withdrawal, under basal conditions and after the resident-intruder paradigm,

by HPLC-EC for male rats. nd- not detectable. Each column represents the mean + S.E.M. of monoamine

levels, expressed as ng of monoamine/mg of protein, for at least 6 animals per group.

nd

Basal Resident-intruder paradigm

saline male cocaine male

245

Prefrontal cortex

A

0

50

100

150

DA DOPAC HVA DA DOPAC HVA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male

Basal Resident-intruder paradigm

B

0

50

100

150

200

250

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male

Resident-intruder paradigmBasal

Figure 4.3.2.2.7- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the prefrontal cortex,

determined 10 days after withdrawal, under basal conditions and after the resident-intruder paradigm, by

HPLC-EC for male rats. Each column represents the mean + S.E.M. of monoamine levels, expressed as ng of

monoamine/mg of protein, for at least 5 animals per group. Columns with the same letter are significantly

different from each other. a p<0.05.

a a

246

Table 4.3.2.2.2- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the dorsal raphe nucleus in saline- and cocaine-treated male, as measured 10 days after

the last administration, under basal condition and after the resident-intruder paradigm.

Basal Resident-intruder paradigm

saline male cocaine male saline male cocaine male

DOPAC/DA 0.89±0.39

(n=6)

0.57±0.24

(n=6)

0.62±0.18

(n=8)

0.53±0.17

(n=9)

HVA/DA 0.10±0.06

(n=6)

0.12±0.04

(n=6) - -

(DOPAC+HVA)/DA 0.99±0.41

(n=6)

0.70±0.27

(n=6)

- -

5-HIAA/5-HT 0.67±0.37

(n=6)

0.87±0.59

(n=6)

0.27±0.06

(n=8)

0.24±0.02

(n=9)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid. In

the same line, values with same letter are significantly different. a p<0.05.

Table 4.3.2.2.3- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the prefrontal cortex in saline- and cocaine-treated male, as measured 10 days after the

last administration, under basal condition and after the resident-intruder paradigm.

Basal Resident-intruder paradigm

saline male cocaine male saline male cocaine male

DOPAC/DA 0.38±0.09

(n=5)

0.31±0.03

(n=6)

0.22±0.06

(n=7)

0.18±0.05

(n=9)

HVA/DA 0.47±0.15

(n=5)

0.49±0.09

(n=6)

0.22±0.07

(n=7)

0.20±0.06

(n=9)

(DOPAC+HVA)/DA 0.85±0.24

(n=5)

0.80±0.09

(n=6)

0.44±0.13

(n=7)

0.38±0.11

(n=9)

5-HIAA/5-HT 0.08±0.02

(n=5)

0.11±0.03

(n=6)

0.06±0.01

(n=7)

0.11±0.03

(n=9)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid. In

the same line, values with same letter are significantly different. a p<0.05, b p<0.02.

Within the hippocampus, cocaine-treated animals present lower levels of DOPAC

after the resident-intruder paradigm, as compared to saline-treated animals [Z=-2.021 (2-

tailed) p<0.05] (figure 4.3.2.2.8). In saline-treated males the levels of DA and 5-HT were

increased after the behavioural test as compared to basal data ([Z=-2.195 (2-tailed)

p<0.05] and [Z=-2.006 (2-tailed) p<0.05], respectively), moreover the 5-HIAA/5-HT

turnover rate was decreased [Z=-2.711 (2-tailed) p<0.01] (table 4.3.2.2.4).

a a

b b

a a

247

Hippocampus

A

0

35

70

105

140

DA DOPAC DA DOPAC

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male

Basal Resident-intruder paradigm

B

0

100

200

300

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male

Resident-intruder paradigmBasal

Figure 4.3.2.2.8- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolite DOPAC and on (B) the levels of 5-HT and its metabolite 5-HIAA in the hippocampus, determined

10 days after withdrawal, under basal conditions and after the resident-intruder paradigm, by HPLC-EC for

male rats. Each column represents the mean + S.E.M. of monoamine levels, expressed as ng of

monoamine/mg of protein, for at least 6 animals per group. Columns with the same letter are significantly

different from each other. a,b,c p<0.05.

a

a

c

c

b b

248

Table 4.3.2.2.4- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA] and [5-HIAA]/[5-HT], in the

hippocampus in saline- and cocaine-treated male, as measured 10 days after the last administration, under

basal condition and after the resident-intruder paradigm.

Basal Resident-intruder paradigm

saline male cocaine male saline male cocaine male

DOPAC/DA 0.24±0.05

(n=6)

0.41±0.11

(n=6)

0.24±0.08

(n=8)

0.29±0.09

(n=9)

5-HIAA/5-HT 0.41±0.07

(n=6)

0.23±0.04

(n=6)

0.14±0.02

(n=8)

0.15±0.02

(n=9)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

Dihydroxyphenylacetic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid. In the same line, values with

same letter are significantly different. a p<0.01.

Within the amygdala, no significant differences were found on the levels of DA, 5-

HT, their metabolites and in turnover rates between cocaine exposed and control males,

after the resident-intruder paradigm (figure 4.3.2.2.9 and table 4.3.2.2.5). However, in

cocaine-treated males the 5-HIAA/5-HT was decreased after the behavioural test as

compared to basal data [Z=-2.326 (2-tailed) p<0.05].

Within the hypothalamus, no significant differences were found on the levels of DA,

5-HT, their metabolites and in turnover rates between cocaine exposed and control males,

after the resident-intruder paradigm (figure 4.3.2.2.10 and table 4.3.2.2.6). However, in

cocaine-treated male rats the levels of DOPAC were decreased after the test as

compared to basal data [Z=-2.333 (2-tailed) p<0.05]. Moreover, cocaine- and saline-

treated males the DOPAC/DA turnover rate was decreased after the behavioural test as

compared to basal data (cocaine-treated males [Z=-2.472 (2-tailed) p<0.02], saline-

treated males [Z=-2.326 (2-tailed) p<0.05]). The 5-HIAA/5-HT turnover rate was also

decrease after the test in cocaine-treated males as compared to cocaine-treated males

under basal conditions males [Z=-2.071 (2-tailed) p<0.05], in the saline-treated males this

difference did not reach significance.

Both in the amygdala and hypothalamus after the behavioural test the levels of

HVA were not detectable by the HPLC-EC.

a a

249

Amygdala

A

DA DOPAC HVA DA DOPAC HVA

0

40

80

120

400

800

1200

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

B

0

50

100

150

200

250

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male

Resident-intruder paradigmBasal

Figure 4.3.2.2.9- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and its

metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the amygdala,

determined 10 days after withdrawal, under basal conditions and after the resident-intruder paradigm, by

HPLC-EC for male rats. Each column represents the mean + S.E.M. of monoamine levels, expressed as ng of

monoamine/mg of protein, for at least 6 animals per group. nd-not detectable.

nd

Basal Resident-intruder paradigm

saline male cocaine male

250

Hypothalamus

A

DA DOPAC HVA DA DOPAC HVA

0

50

100

600

1200

1800

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

B

0

125

250

375

500

5-HT 5-HIAA 5-HT 5-HIAA

Neu

rotr

ansm

itter

s co

ncen

trat

ion

(ng/

mg

of p

rote

in)

saline male cocaine male

Resident-intruder paradigmBasal

Figure 4.3.2.2.10- Effects of chronic cocaine exposure throughout adolescence on (A) the levels of DA, and

its metabolites, DOPAC and HVA and on (B) the levels of 5-HT and its metabolite 5-HIAA in the

hypothalamus, determined 10 days after withdrawal, under basal conditions and after the resident-intruder

paradigm, by HPLC-EC for male rats. Each column represents the mean + S.E.M. of monoamine levels,

expressed as ng of monoamine/mg of protein, for at least 5 animals per group. nd- not detectable. Columns

with the same letter are significantly different from each other. a p<0.05.

nd

a a

Basal Resident-intruder paradigm

saline male cocaine male

251

Table 4.3.2.2.5- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the amygdala in saline- and cocaine-treated male, as measured 10 days after the last

administration, under basal condition and after the resident-intruder paradigm.

Basal Resident-intruder paradigm

saline male cocaine male saline male cocaine male

DOPAC/DA 0.33±0.19

(n=6)

0.34±0.13

(n=6)

0.16±0.06

(n=7)

0.12±0.04

(n=8)

HVA/DA 0.08±0.04

(n=6)

0.08±0.02

(n=6) - -

(DOPAC+HVA)/DA 0.41±0.19

(n=6)

0.42±0.13

(n=6) -

-

5-HIAA/5-HT 0.30±0.09

(n=6)

0.47±0.15

(n=6)

0.20±0.03

(n=7)

0.15±0.02

(n=8)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid. In

the same line, values with same letter are significantly different. a p<0.05.

Table 4.3.2.2.6- Turnover of DA and 5-HT, expressed as [DOPAC]/[DA], [HVA]/[DA], [DOPAC+HVA]/[DA] and

[5-HIAA]/[5-HT], in the hypothalamus in saline- and cocaine-treated male, as measured 10 days after the last

administration, under basal condition and after the resident-intruder paradigm.

Basal Resident-intruder paradigm

saline male cocaine male saline male cocaine male

DOPAC/DA 0.43±0.12

(n=6)

0.41±0.09

(n=5)

0.14±0.03

(n=8)

0.11±0.03

(n=9)

HVA/DA 0.03±0.01

(n=6)

0.07±0.03

(n=5) - -

(DOPAC+HVA)/DA 0.45±0.12

(n=6)

0.48±0.11

(n=5) -

-

5-HIAA/5-HT 0.31±0.09

(n=6)

0.37±0.10

(n=5)

0.15±0.02

(n=8)

0.17±0.03

(n=9)

Data expressed as the mean value ± S.E.M.; n, number of animals per group. DA- dopamine; DOPAC- 3,4-

dihydroxyphenylacetic acid; HVA- homovanillic acid; 5-HT- serotonin; 5-HIAA- 5-hydroxyindolacetic acid. In

the same line, values with same letter are significantly different. a,c p<0.05, b p<0.02.

a a b b

c c

a a

252

4.4. Discussion

4.4.1. Effects of chronic cocaine treatment during adolescence and withdrawal on the body weight evolution

The data on body weight was analyzed in terms of body weight gain, and during

the experimental period no effects of cocaine treatment were observed. These data

contradict the known anorexic effects of cocaine (Spector et al., 1972; Balopole et al.,

1979; Cooper and van der Hoek, 1993). However, other authors had already reported no

effects of cocaine administration in body weight evolution in adolescent rats (Laviola et al.,

1995; Giorgetti and Zhdanova, 2000). As already discussed in chapter 3, the absence of

cocaine effects on body weight evolution may be related with a strategy adopted during

adolescence, where the available resources are directed into ponderal growth, whereas in

the adult resources are preferentially used to maintain the reproductive function (Gomez

and Dallman, 2001; Gomez et al., 2002). Nevertheless, it was observed a sex effect in the

body weight evolution, male rats had higher weight gain than females. This effect was

expected, since sex differences in body weight appear during adolescence (Macri et al.,

2002).

4.4.2. Effects of chronic cocaine treatment during adolescence and withdrawal on the neuroendocrine function

As already discussed in chapter 3, cocaine has been reported to have effects on

the HPA axis activity. This work aimed to study the effects of chronic cocaine exposure

during adolescence on HPA axis activity throughout long-term withdrawal. Two time points

of assessment were chosen, 5 (an assessment point during adolescence period) and 10

days (beginning of adulthood) after the last cocaine administration.

It has been demonstrated that the HPA axis activity was disrupted (Sarnyai et al.,

1998; Mantsch et al., 2000) during withdrawal from chronic cocaine treatment, suggesting

that altered HPA axis function may play an important role in cocaine addiction. In fact,

acute elevation of glucocorticoids has been reported to precipitate reinstatement in rats

(Deroche et al., 1997) and craving in humans (Elman et al., 2003). Also in rats,

adrenalectomy prevents reinstatement of self-administration behaviour following extinction

(Erb et al., 1998). Elevated plasma corticosterone appears to contribute to the stress-

induced reinstatement of extinguished cocaine-seeking behaviour in animal models of

drug relapse (Deroche et al., 1997; Mantsch and Goeders, 1999). These findings indicate

that corticosterone is critical for the reinstatement of extinguished cocaine-seeking

behaviour, demonstrating an involvement of the HPA axis in the relapse to cocaine use.

253

Our data in chronic binge exposure to cocaine during adolescence indicate that

both 5 or 10 days after cocaine withdrawal the basal levels of ACTH and corticosterone in

male and female rats did not significantly differ from those of the control animals. These

results may confirm previously findings indicating that chronic cocaine administration does

not seem to have prolonged effects upon the HPA axis activity under basal conditions.

Teoh at al. (1994) found no significant differences in pulsatile release of ACTH in men

with a past history of cocaine abuse who were studied under controlled drug-free

conditions in comparison to normal controls (Teoh et al., 1994). After a brief period of

abstinence, basal and CRH-stimulated ACTH and cortisol levels in cocaine-dependent

patients were not different from healthy subjects (Vescovi et al., 1992; Baumann et al.,

1995; Jacobsen et al., 2001). In rats, Zhou et al. (2003), using a exposure paradigm

similar to the one used in the present study, showed that plasma ACTH and

corticosterone levels remained increased 4 days after withdrawal, but 10 days later were

at control levels. Rats chronically treated with cocaine (15mg/kg i.p.) for 14 days and then

challenged with an acute dose of cocaine 7 days after the last chronic cocaine treatment

displayed ACTH and corticosterone responses that did not differ from those under control

treatment conditions (Levy et al., 1992a). These findings were confirmed and extended to

periadolescent rats, after 4 days of cocaine administration (0,10 or 20 mg/kg i.p.) a

challenge dose of cocaine (10 mg/kg ip) had equivalent effects on ACTH and

corticosterone in the control and cocaine-treated groups (Laviola et al., 1995).

Nevertheless, our results showed that although no differences were observed in

corticosterone levels between groups at the same time point, saline-treated females

displayed an increase in ACTH levels from 5 to 10 days after withdrawal, with this effect

not being observed in cocaine-treated females. In males, this effects is observed in

cocaine-treated males (although no significantly) and not in control males. This may

indicate an effect of the cocaine treatment in the function of the HPA axis after withdrawal;

moreover this effect is sex-dependent.

In the present study the analysis of the effects of cocaine withdrawal on HPA axis

activity was extended besides the hormonal measures and the levels of CRH mRNA in

hypothalamus and the levels of POMC and CRH-R1 mRNAs in the pituitary were

analyzed in male rats 10 days after the end of the exposure period. The results did not

showed significant differences in the levels of expression of the analyzed mRNAs both in

hypothalamus and pituitary between cocaine-treated and control males. These measures

were only conducted for male rats, as justified in page 215.

It is known that a single injection of cocaine, or an acute binge pattern cocaine

administration elevates CRH mRNA levels in the hypothalamus in rats (Rivier and Lee,

254

1994; Zhou et al., 1996b), with no significant alterations in either CRH-R1 or POMC

mRNA levels in the anterior pituitary (Zhou et al., 1996b), indicating the importance of the

hypothalamic CRH in modulating the stimulatory effects of the HPA axis. However, Zhou

at al. (2003) showed that 1 day after a period of 14 days of chronic binge cocaine

administration did not lead to an increase in CRH mRNA levels in the hypothalamus (Zhou

et al., 2003).

CRH-R1 mRNA expression in the anterior pituitary appears to be preferentially

associated with ACTH-positive cells, and both POMC and CRH-R1 mRNA expression are

subject to negative feedback control by glucocorticoids (e.g., Zhou et al., 1996a). The

study by Zhou et al. (2003) also showed that both of the POMC and CRH-R1 mRNA

expression levels were increase 1 day after chronic binge cocaine administration but

returned to control after 4 days into withdrawal. In another study it was reported that

prolonged withdrawal from cocaine was associated with normalization of regional brain

CRH synthesis (Zhou et al., 1996b) and CRH receptor binding (Ambrosio et al., 1997) by

10 days post-withdrawal. Those data indicate that chronic cocaine binge administration

does not seem to have prolonged effects upon the HPA axis activity under basal

conditions. Our findings seems to confirm and extend this data to adolescent rats.

In the present study we also investigated the feedback inhibitory mechanisms, by

examining GR mRNA expression levels in the pituitary and brain regions putatively

involved in feedback regulation of the HPA axis. No differences were observed between

cocaine- and saline-withdrawn male rats. Although, it cannot be rule out the possibility that

GR affinity or function was altered without changes in mRNA expression. These data also

seems to indicate an absence of prolonged effects of cocaine treatment upon the HPA

axis activity under basal conditions.

Data from adrenal weight was collected 5 and 10 days after the withdrawal. The

weights were normalized to body weight and data analysis revealed that adrenal weights

were not affected by the cocaine treatment during adolescence since no differences were

observed between cocaine-treated males and females and their respective controls at

both time points of assessment. Nevertheless, and as already observed 30 minutes and 1

day after the last administration of cocaine, and discussed in chapter 3, there was a sex

effect on the adrenal weight, with adrenals of males weighing significantly less than that of

the females, which is in agreement with published literature (Majchrzak and Malendowicz,

1983).

In the present study the long-term effects of chronic cocaine exposure during

adolescence on gonadal steroid levels were examined, both in male and female rats, 5

255

and 10 days after cocaine withdrawal. As adolescence includes puberty, a period of

marked hormonal changes, external perturbation of this developmental process may lead

to long-term effects on the HPG axis function.

It has been difficult to detect changes in basal hormone levels after chronic

cocaine abuse in animal models. However, it has been infered that chronic cocaine

changes in neuroendocrine function do occur on the basis of clinical and experimental

evidence of cocaine-related disruption of the reproductive function (Siegel, 1982; Cocores

et al., 1986; Berul and Harclerode, 1989; Chen and Vandenbergh, 1994).

Our data showed that 5 days after cocaine withdrawal plasma levels of

testosterone did not differ between chronically cocaine- and saline-treated male rats.

However, saline-treated males presented an increase in levels of testosterone from 5 to

10 days; this effect was not observed in cocaine-treated males. In fact, cocaine-treated

males displayed lower levels of testosterone at 10 days after cocaine withdrawal as

compared to control males. As discussed in chapter 3, acute cocaine administration is

known to induce significantly decreases in plasma levels of testosterone (Berul and

Harclerode, 1989). The decreased testosterone plasma levels observed 10 days after

withdrawal, a time point that matches the beginning of adulthood, may be a consequence

of repeated decrease in testosterone levels after each cocaine administration throughout

the exposure period on the development of testes occurring during this period. In

agreement with this hypothesis, chronically treated males also presented decreased

relative gonadal weights at this time point, which may suggest that the observed

decreased testosterone levels may be primarily related with a reduced steroidogenic

capacity of testes, probably due to an abnormal maturation of the gonads. Other authors

have shown that morphological changes and lesions in testes are present in the rat after a

long-term exposure to cocaine. Rats long-term exposed to cocaine, starting on

peripubertal stage, presented reduced mean diameter of seminiferous tubules, thiner

germinal epithelium, and numerous degenerating cells, although in that work the levels of

testosterone were unaltered (George et al., 1996). In adult rats, Barroso-Moguel et al.

(1994), showed that in long-term exposed animals, testes were diminished in size, the

seminiferous tubules were shrunken, had small diameter and contained necrotic

spermatogenic cells and fibrosis.

In the present study no differences were found in the progesterone plasma levels

between cocaine- and saline-treated males, as accessed 5 and 10 days after cocaine

withdrawal. The fact that testosterone, but not progesterone plasma levels were reduced

at 10 days after cocaine withdrawal suggest that cocaine may interfere with some specific

metabolic processes in the synthesis of testosterone in Leydig cells. Although LH levels

256

were not measured, our data seems to strongly reflect a direct effect of cocaine on the

testes.

The study of the long-term effects of withdrawal from chronic cocaine treatment

throughout adolescence on the HPG axis activity of male rats was extended besides the

hormonal level determinations and the expression levels of GnRH-R, LHβ and AR mRNAs

in pituitary, and levels of AR mRNA in hypothalamus were analyzed at 10 days after

cocaine withdrawal. No differences were observed between cocaine- and saline-

withdrawn male rats. The absence of alterations on expression levels of AR mRNA seems

to indicate that the testosterone negative feedback mechanism at the pituitary and the

hypothalamus was not altered 10 days of withdrawal. Data on GnRH-R mRNA expression

levels seems to indicate that pituitary responsiveness to hypothalamic GnRH pulses was

not altered; moreover the LHβ mRNA levels were also unaltered as compared to control

animals. These data appears to support the hypothesis that reduced levels of testosterone

observed in cocaine-treated males result primarily from impaired testicular

steroidogenesis.

Little is known about the effects of cocaine on the endocrinological profile of

females, moreover the long-term effects have been almost unexplored. Nevertheless, the

persistent menstrual cycle abnormalities observed during cocaine withdrawal suggest that

the disruptive effects on the menstrual cycle do not only reflect an acute intoxication

outcome (Mello et al., 1997). Rather, daily cocaine exposure may induce long-lasting

disruptions of the neuroendocrine regulation of the menstrual cycle. Persistent effects of

cocaine after chronic exposure have been observed in several models (Mello and

Mendelson, 1997). In rats, estrous cycles did not return to normal after high doses of

cocaine over 5 to 6 weeks of observation (King et al., 1993b).

Our data showed that females chronically exposed to cocaine throughout

adolescence presented higher levels of progesterone at 10 days after the last cocaine

administration. These high levels of progesterone may inhibit the release of LH preventing

ovulation, leading to a disruption of the normal estrous cycle hormone regulation following

chronic cocaine administration. Moreover, saline-treated females displayed decreased

plasma levels of progesterone between 5 and 10 days after the ending of the exposure

protocol, with this effect not being observed in cocaine-treated females. In fact,

Feltenstein and See (2007) had already showed increased progesterone levels during and

after cocaine self-administration in the non-estrus phases of the cycle (Feltenstein and

See, 2007). However, Walker et al. (2001) reported that the cocaine-induced an increase

in plasma progesterone levels, which was not present in adrenalectomized animals,

257

suggesting that this effect may involve an action of cocaine on the HPA axis activity, either

through adrenally derived progesterone release or HPA axis/ovary interactions.

No differences in the plasma levels of estradiol were observed between cocaine-

and saline-treated females. Nevertheless, the levels of estradiol in saline-treated females

increased from 5 to 10 days after the last administration, with this effect not being

observed in cocaine-treated females, supporting an impaired regulation of the HPG axis

during the withdrawal period.

While suggestive, the results described here do not consider the physiological

fluctuations of ovarian hormones already present in adolescent females. Although at this

age it difficult to determine the stage of estrous cycle, this can interfere with the present

results.

Overall, our results indicate a long-lasting disruptive effect of cocaine on the HPG

axis function in male, and a trend to a long-term impaired regulation of the HPG axis in

female rats.

4.4.3. Effects of chronic cocaine treatment during adolescence on the dopaminergic and serotonergic systems function

Recently, there has been renewed interest in the neuroadaptation induced during

periods of cocaine withdrawal. Moreover, there are conceptual reasons for expecting that

drug consumption may affect adolescents differently than adult consumption. Few studies

have investigated whether the continued plasticity during this late development extends to

drug-induced changes in the CNS, although several reports indicate that cocaine (Smith

et al., 1999; Estelles et al., 2007) and alcohol (Siciliano and Smith, 2001) during

adolescence may induce some long-lasting behavioural changes. For example, Catlow

and Kirstein (2007) demonstrated that in rats pretreated with cocaine during the

adolescence there is an enhanced response of the dopaminergic system to naturally

reinforcing substances, suggesting long-term functional changes in the mesolimbic

pathway (Catlow and Kirstein, 2007). Brandon and colleagues administered 15mg/kg of

cocaine for 5 days and found that adolescent rats pretreated with cocaine showed

persistent behavioural sensitization two months after cocaine cessation (Brandon et al.,

2001). The use of cocaine during adolescence could alter the normal growth of brain

regions affected by cocaine, specifically the reward system, and impact the adult

mesolimbic system. However, there is limited literature aiming to determining whether

animals are more vulnerable to the adverse effects of drugs during adolescence (Catlow

and Kirstein, 2007). Thus, in the present study, the objective of investigate whether

258

cocaine pretreatment in adolescence altered the dopaminergic or serotonergic systems

activity at 5 or 10 (early adulthood) days after withdrawal, was also established.

- Effects on the mesoaccumbens and nigrostriatal pathways

Our data showed that, 5 days after the last administration adolescent rats treated

chronically with cocaine presented no differences on the levels of DA and its metabolites,

DOPAC and HVA, in regions expected to be affected by cocaine exposure, such as the

SN-VTA, the Nac and the dorsal striatum, as compared to saline controls. Several studies

indicate that alterations in the activity of midbrain DA neurons during withdrawal are

dependent on the type of cocaine administration protocol that is used. Whereas

intermittent cocaine injections have been shown to increase extracellular DA

concentrations in the striatum during withdrawal, continuous cocaine infusions produced

the opposite effect (King et al., 1993a). Similarly, withdrawal from continuous cocaine

administration decreased the number of spontaneously active midbrain DA neurons,

whereas withdrawal from an intermittent schedule of cocaine increased the number of

spontaneously active DA neurons (Gao et al., 1998). Considering the relatively high

dosing delivered in the binge cocaine administration experiments, it was expected that the

findings for chronic binge administration were similar to those observed in rats during

withdrawal from continuous cocaine administration, thus it was expected to find a

depression of the dopaminergic system activity.

However, the absence of differences in the activity of the dopaminergic system

between saline- and cocaine-treated animals observed in the present study may result of

increased dopaminergic activity induced by the expectancy of drug administration in

cocaine-treated animals, since animals were decapitated in the same room where the

cocaine was daily administered. As already discussed in chapter 3, several investigators

have demonstrated a stimulation of dopaminergic activity when drug-free rats were

exposed to environments that have been previously paired with stimulant drugs, such as

amphetamine or cocaine (i.e. a conditioning effect) (Post et al., 1981; Badiani et al., 1995;

Antkiewicz-Michaluk et al., 2006). In the present study, throughout the period of

withdrawal, the animals were brought daily to the room where cocaine was administered,

without receiving any injection, in order to eliminate the expectancy of drug administration,

and thus eliminate the activation of dopaminergic system induced by the cocaine cues

presentation. However, based in our data obtained at 5 days after withdrawal, we

speculated that by this time the expectancy of drug administration was still present.

259

Ten days after cocaine withdrawal our data seems to point toward an increase in

dopaminergic activity in male rats, as indicated by the increased DOPAC levels in SN-

VTA and the increased levels of HVA in dorsal striatum. In fact, the mRNA expression

levels of tyrosine hydroxylase (TH) were increased in SN-VTA 10 days after withdrawal in

male rats. TH is the key enzyme for synthesizing DA in dopaminergic neurons and its

terminals. We speculate that the increase in TH in the SN-VTA may reflect a

compensatory response of dopaminergic neurons to upregulate their synthesizing

capacity. This data supports the findings from neurochemical analyses, showing that

chronic cocaine administration throughout the adolescence period has long-lasting effects

upon the dopaminergic system. Moreover, these results suggest an increase in the DA

synthesis, likely to compensate for a chronic cocaine-induced depression of the

dopaminergic system. Other authors have reported alteration in the dopaminergic system

activity at 10 days withdrawal although in the opposite direction. Antkiewicz-Michaluk and

colleagues (2006) reported that 10 days of withdrawal from self-administered cocaine

leads to an impairment of the function of the dopaminergic system, as reflected by a

significant decline in DA concentration in the Nac and striatum, but not in FC, SN and VTA

(Antkiewicz-Michaluk et al., 2006). There are two factors that vary between the latter

mentioned and the present study that could be involved in the different results obtained:

the cocaine dosing regime (i.v. self-administration versus i.p. experimenter-delivery) and

the period of exposure (adulthood versus adolescence).

It was already reported that basal levels of DA in the shell region of the Nac were

affected differentially as a function of age by repeated cocaine use (Catlow and Kirstein,

2007). Specifically, rats pretreated with cocaine in the adulthood had lower baseline levels

of DA than saline-pretreated rats. It has been postulated that a decrease in the levels of

TH within the Nac leads to decreased basal levels of DA, and this results in a postsynaptic

downregulation of the dopaminergic system (Parsons et al., 1991; Segal and Kuczenski,

1992; Maisonneuve et al., 1995). It is interesting that rats pretreated in the adolescence

with the same dose of cocaine for the same length of time did not have a reduction in

basal DA levels (Catlow and Kirstein, 2007). These results suggest that the administration

of cocaine differentially induces long-term changes in the dopaminergic transmission in

adolescent and adult rats.

Due to the increasing numbers of DATs through adolescence, higher reuptake will

decrease basal DA in the striatum (Andersen and Gazzara, 1993) and in the Nac (Philpot

and Kirstein, 1999) and upregulates cyclic-AMP (cAMP) signaling (Andersen, 2002). The

profile of the adolescent mesolimbic system, with lower basal levels of DA, increased DA

receptors and cAMP levels, could be overstimulated in the presence of cocaine.

260

Persistent postsynaptic stimulation hyperpolarizes GABAergic neurons and allows an

increased release of DA into the extracellular space which could in part explain cocaine-

induced hyperactivity observed in adolescent rats (Catlow and Kirstein, 2005; Maldonado

and Kirstein, 2005b, a). During adolescence, glutamatergic projections from prefrontal

cortex to VTA and limbic areas develop (reviewed by Lewis, 1997). It is plausible that

repeated cocaine during adolescence overstimulates the mesolimbic system causing

more synaptic connections of glutamatergic afferents to the VTA. Repeated cocaine

during adolescence could result in a circuitry primed for vulnerability to addiction.

Our results seem to indicate that the nigrostriatal system of the in male rats is the

most affected dopaminergic pathway by the cocaine treatment. The striatum is thought to

have a role in motor activity, such as stereotypes (Creese and Iversen, 1974; Robbins and

Everitt, 1996), but also in rewarded “habit” learning (Everitt and Wolf, 2002), so the

present results suggest that the observed neurochemical changes may be partially

responsible for withdrawal symptoms. Moreover, the increased dopaminergic activity

observed in male rats after long-term withdrawal may render the individuals more

susceptible to the reward effects of cocaine, therefore more vulnerable to reinstatement.

In most dopaminergic cells, DAT mRNA and TH mRNA-positive neurons

completely overlap (Hoffman et al., 1998). This co-localization may be due to similar

molecular mechanisms of transcriptional regulation of these genes. The data descried in

the present study indicate that expression levels of DAT mRNA, in male rats 10 days after

withdrawal, were increased in SN-VTA in a way that almost reached significance.

Whereas acute administration of cocaine did not alter DAT mRNA levels in the VTA and

SN (Xia et al., 1992; Spangler et al., 1996), they were reported to be decreased by

chronic cocaine treatment (Xia et al., 1992; Burchett and Bannon, 1997; Letchworth et al.,

1997; Letchworth et al., 1999). Following withdrawal from cocaine, DAT mRNA expression

was unaltered in the SN at 72 hours withdrawal period (Xia et al., 1992), but decreased in

parts of the VTA at 10 days after 2 weeks i.v. cocaine treatment (Cerruti et al., 1994).

Maggos et al. (1997), using the same pattern of cocaine administration as those used in

the present study, reported unchanged DAT mRNA levels in both areas under subacute (3

days) binge, chronic (14 days) binge or 10 days withdrawal from a chronic binge pattern

cocaine. However, we did observe a trend to increased expression levels of DAT mRNA

in the SN-VTA in the cocaine-treated male rats 10 days after withdrawal. Arroyo et al.

(2000) also reported a long lasting up-regulation of the DAT mRNA levels in the VTA 10

days after animals had self-administered cocaine for 10 days. This data seems to support

that upregulation of DAT mRNA induced by cocaine administration may depend on the

period of cocaine exposure more than on the cocaine dosing regime. The upregulation of

261

the gene encoding DAT could be the result of a compensatory mechanism that develops

due to the repeated DAT blockade-induced increases in extracellular DA levels following

cocaine exposure.

Nevertheless, some studies have shown no correlation between cocaine-induced

effects on DAT mRNA level and concentration and binding site densities at the synaptic

terminals, when evaluated at the same time point after non-contingent cocaine treatment

(Cerruti et al., 1994; Letchworth et al., 1997; Letchworth et al., 1999). This may reflect the

ability of cocaine to alter some of the molecular processes involved in protein synthesis

(i.e. rates of transcription, translation, mRNA degradation, axonal transport, protein

stability).

The expression levels of DA receptors D1, D2 and D3 mRNA were not altered 10

days after withdrawal in cocaine exposed male rats. There is considerable evidence that

chronic exposure to cocaine is associated with low striatal D2 receptor availability.

Moreover, imaging studies using PET have demonstrated that reduced striatal D2

receptor availability in functional subdivisions of the striatum, might be involved in the

initiation and/or maintenance of cocaine abuse. Indeed, low striatal D2 receptor availability

was observed in detoxified chronic cocaine abusers (Volkow et al., 1993), and in healthy

subjects was consider as predictive of a pleasurable experience following administration

of the psychostimulant methylphenidate (Volkow et al., 1999). A recent study by Stefanski

and colleagues (2007) reported that passively administered cocaine produced a decrease

in D2 receptor levels in the anterior and central regions of caudate-putamen, and the Nac, as measured by in vitro quantitative autoradiography. On the other hand, they reported

that self-administration of cocaine increased D2 receptor mRNA levels in the VTA,

although with no corresponding differences in D2 binding density (Stefanski et al., 2007).

This later result are consistent with previous studies demonstrating that self- administred

cocaine had no effect on D2 receptor density in the dorsal and ventral striatum of rats

(Laurier et al., 1994). Importantly, these results suggest that the reductions in striatal D2

receptor densities may be related to the direct pharmacological actions of chronic

administration of cocaine per se and not to the motivated process of reinforced

responding.

A drug-free period of 7 days after repeated intermittent treatment with cocaine for

14 days was reported to result in a functional subsensitivity of release-modulating D2

autoreceptors in the caudate (Jones et al., 1996) and in the Nac of rats (Davidson et al.,

2000). However, Svensson and Hurd (1998) reported that rats treated with cocaine (30

mg/kg, i.p. once daily) for 10 days, followed by a 10-day drug free-period displayed

262

decreased expression levels of D1-receptor mRNA in the caudorostral striatum and

unaltered expression of D2-receptor mRNAs (Svensson and Hurd, 1998).

It has been reported that treatments inducing behavioural sensitization to cocaine,

all of which used experimenter administered i.p. injections, produce no change in the total

levels of D1 or D2 receptor in the VTA, Nac, striatum, or prefrontal cortex (for reviews, see

Pierce and Kalivas, 1997; Vanderschuren and Kalivas, 2000). These data agree with the

results obtained in the present study, showing no differences in the expression levels of

DA receptors mRNAs in cocaine-treated male rats 10 days after withdrawal as compared

to controls. In fact, the paradigm of drug administration used in the present study is known

to induced behavioural sensitization (Lucas et al., 1997; Caster et al., 2005).

The data obtained in cocaine-treated females, during the drug free-period point to

the absence of treatment effects in the dopaminergic neurotransmission within the SN-

VTA, Nac and dorsal striatum, again diverging from the data obtained in male rats. There

are growing evidences that females display a differential response and adaptation to

stress than do males (Dalla et al., 2008; Lin et al., 2009; also see for review Ter Horst et

al., 2009). Thus, the high levels of stress that animals were submitted into, particularly the

stress caused by the social isolation, could have masked the effects of cocaine and its

withdrawal in female rats.

Preclinical and clinical findings showed that withdrawal from chronic cocaine is

associated with impaired 5-HT neuronal function in addition to the well-accepted deficits in

DA function (Dackis and Gold, 1985). Perhaps the most compelling evidence of 5-HT

deficits in cocaine addiction is the occurrence of a psychiatric syndrome resembling major

depression that follows abstinence from binge cocaine use (Dackis and Gold, 1985;

Gawin and Kleber, 1986), coupled with the increased prevalence of suicidal ideation and

suicide attempts among cocaine addicts (Garlow et al., 2003). The established role of 5-

HT dysfunction in mediating depression and suicide (Mann, 2003) indicates that

decreased synaptic 5-HT levels could have a role in cocaine withdrawal states (Baumann

and Rothman, 1998). In addiction, increasing postsynaptic DA function does not reliably

reduce cocaine craving (Kampman et al., 1996; Handelsman et al., 1997). However,

promising results have been obtained for drugs that increase extracellular 5-HT levels

(Pollack and Rosenbaum, 1991; Batki et al., 1993; Kampman et al., 2000) or act directly

on 5-HT receptors (Buydens-Branchey et al., 1997; Buydens-Branchey et al., 1998).

In vivo microdialysis experiments reveal that rats withdrawn from i.v. cocaine self-

administration have decreased extracellular levels of 5-HT in the Nac (Parsons et al.,

1995). A feasible mechanism to explain this finding is, just like described for DA (Wilson et

263

al., 1994; Mash et al., 2002), the enhanced uptake of 5-HT. Indeed, chronic cocaine

administration can increase the density of SERTs in rats (Cunningham et al., 1992) and

humans (Jacobsen et al., 2000). Increases in SERT density in the rostral ventromedial

caudate putamen, Nac shell, and olfactory tubercle were demonstrated in adult rats

pretreated with cocaine (Collins and Izenwasser, 2002). However, no significant changes

in SERT density were observed in periadolescent rats 10 days after withdrawal compared

to vehicle controls, showing that periadolescent rats have different serotonergic

adaptations to repeated cocaine administration than adult rats (Collins and Izenwasser,

2002).

In the present study neurochemical data did not showed a decrease in 5-HT levels

in cocaine-treated animals as compared to controls. However, the levels of 5-HT

increased from day 5 to day 10 after cocaine withdrawal in male rats in the SN-VTA, while

in cocaine-treated female rats the levels of 5-HT and 5-HIAA increased from 5 to 10 days

after withdrawal in the Nac. These effects were not observed in saline controls, indicating

that in fact cocaine treatment during adolescence have long-lasting effects on the

serotonergic activity, and like as for the dopaminergic activity, the increase in 5-HT levels

may represent an attempt to counteract the downregulation of the serotonergic system

activity during withdrawal.

- Effects on the prefrontal cortex

Cocaine addicts have a number of cognitive deficits that persist following

prolonged abstinence. These include impaired performance on tasks involving attention

and cognitive flexibility that are thought to be mediated by the medial and orbital prefrontal

cortex, as well as spatial, verbal, and recognition memory impairments on tasks thought to

be mediated by the hippocampus (Manschreck et al., 1990; Ardila et al., 1991; Berry et

al., 1993; Beatty et al., 1995; Hoff et al., 1996; Bolla et al., 2003).

DA is an essential contributor within the prefrontal cortex during cocaine abuse

(Kelley, 2004). DA is a critical modulator of the pyramidal neuron excitability and excitatory

synaptic transmission in the prefrontal cortex (Seamans and Yang, 2004; Floresco and

Tse, 2007). Pyramidal neurons in the medial prefrontal cortex and their afferents express

receptors for DA (Smiley et al., 1994; Krimer et al., 1997). It has been reported that after

chronic cocaine administration, dopaminergic neurotransmission in the prefrontal cortex

undergoes significant changes (Williams and Steketee, 2005), time-dependent changes

that may be caused by alteration in binding to DA receptor subtypes (Ben-Shahar et al.,

2007).

264

Our study on the prefrontal cortex dopaminergic activity indicates that the levels of

DA and its metabolites DOPAC and HVA did not differ between cocaine- and saline-

treated animals, both at 5 and 10 days after withdrawal. Other studies have also revealed

that withdrawal from repeated cocaine produce no statistically significant changes in

baseline DA levels within the prefrontal cortex (Sorg et al., 1997; Chefer et al., 2000;

Williams and Steketee, 2005). However, for instance, repeated systemic administration of

cocaine produced time-dependent alterations in cocaine-induced increases in extracellular

DA levels within the medial prefrontal cortex (Williams and Steketee, 2005). In that study,

1 day after withdrawal from repeated cocaine the dopaminergic response to an acute

cocaine challenge was blunted in cocaine-pretreated animals as compared to controls,

with this attenuated response being more pronounced when examined after 7 day of

withdrawal; while after 30 days of withdrawal it was observed an augmented medial

prefrontal cortex DA response to cocaine, in cocaine-treated rats (Williams and Steketee,

2005). These results suggest that withdrawal from repeated cocaine is associated with

marked alterations in prefrontal cortex DA neurotransmission. However, in our study,

beside the absence of cocaine treatment effects revealed by the neurochemical data

obtained 5 and 10 days after withdrawal, the analysis of expression levels of D1, D2 and

D3 receptors, DAT and MAO-A mRNAs, in the prefrontal cortex of male rats, 10 days after

withdrawal, also revealed no significant differences between cocaine- and saline-treated

male rats. Nogueira et al. (2006) also reported no alterations on the effects of D1 receptor

antagonists on the current-evoked and spontaneous excitability of pyramidal cells at 2-4

weeks after a repeated cocaine treatment of 7 days, while reporting that D2 receptor-

mediated regulation of cortical excitability was reduced. After long-access cocaine self-

administration was also reported a significant decrease in D2 (but not D1) mRNA in the

medial and orbital prefrontal cortex, and D2 receptor protein in the medial prefrontal cortex

of rats (Briand et al., 2008) An extensive body of evidence describes the fundamental role

of prefrontal cortex D2 receptors in cocaine addiction. Beyer and Steketee (2002)

demonstrated that intra-medial prefrontal cortex injections of the D2 receptor agonist

quinpirole blocked cocaine-induced sensitization (Beyer and Steketee, 2002). Moreover,

in studies with human addicts, Volkow et al. (2001) reported lower levels of D2 receptor

availability in the orbitofrontal cortex, suggesting that dysregulation of this type of receptor

could underlie the loss of control and compulsive drug intake in drug addicts. The

contradictory findings between the above mentioned studies and our study could be

related with the different dosing regimen exposure and the period of exposure. In contrast

to the other studies that used adult animals, in our study, were used adolescent animals

undergoing prominent developmental transformations (Spear, 2000). Therefore, we

propose that the developmental processes may underlyie the differences observed

265

between our study and these carried out in adults. It also important to note that, although

no differences were observed in mRNA expression levels this, does not exclude the

possibility that the DA receptors concentration and binding site densities could have been

altered.

- Effects on the hippocampus

In the hippocampus, cocaine-treated males did not present alterations in levels of

DA or DOPAC both 5 and 10 days after cocaine withdrawal, with the DA turnover rate also

not being significantly different from that of the control animals. The expression levels of

D1, D2 and D3 receptors, DAT and MAO-A mRNA, determined 10 days after withdrawal

in male rats, also did not revealed any treatment effects. Together these data seems to

suggest that basal dopaminergic neurotransmission in the hippocampus was not long-

term affected by the chronic binge cocaine exposure throughout adolescence in the male

rats. However, in cocaine-treated females the levels of DA increased from 5 to 10 days

after withdrawal, while this effect was not observed in control females, suggesting a

different pattern of evolution in the dopaminergic neurotransmission between cocaine-

treated and control females. The pivotal role of the hippocampal dopaminergic system has

been demonstrated in several types of learning: intrahippocampal injections of DA

agonists enhance passive avoidance (Bernabeu et al., 1997) and win-shift positive

reinforcement learning (Packard and White, 1991). DA depletion in the hippocampus

impairs spatial navigation (Gasbarri et al., 1996). It is known that one form of synaptic

plasticity, long-term potentiation (LTP) at the CA1 Schaffer collateral synapses, is

facilitated by D1/D5 DA receptors (Frey et al., 1990; Huang and Kandel, 1995; Otmakhova

and Lisman, 1996). Thus alterations in the dopaminergic neurotransmission induced by

pre-exposure to cocaine may interfere with processes of LTP and learning/memory

formation.

Besides the observed alteration in hippocampal dopaminergic function in the

cocaine-treated female rats during withdrawal, the serotonergic neurotransmission in this

brain formation also seems to be affected by cocaine treatment. Saline-treated males

presented an increase in the 5-HIAA/5-HT turnover rate from 5 to 10 days after the last

administration, suggesting an increase in the 5-HT metabolism that was not observed in

the cocaine-treated males, and therefore, a cocaine treatment effect in the serotonergic

pattern of evolution throughout withdrawal. In cocaine-treated females, the levels of 5-

HIAA increased from 5 to 10 days after withdrawal, as well as it was observed a trend for

an enhanced 5-HIAA/5-HT turnover rate, suggesting an increase in the 5-HT metabolism

266

throughout long-term withdrawal. These data point to an altered hippocampal function

after long-term cocaine withdrawal, both in male and female rats cocaine exposed during

adolescence, Kimura et al. (2004) also reported changes in hippocampal serotonergic

modulation several days after cessation of repeated administration of methamphetamine,

even though, 10 days after withdrawal, these changes were no longer observed. Reports

on the long-term effects of cocaine on hippocampal serotonergic activity are scarce.

Nevertheless, for instance Przegalinski and colleagues (2003) reported that withdrawal

from chronic cocaine induces up-regulation of 5-HT1B receptors (Przegalinski et al.,

2003). Some evidence indicates that up-regulation of the 5-HT1B receptors occurs

following impairment of 5-HT neurotransmission, e.g. after 5-HT denervation with 5,7-di-

hydroxytryptamine, or after inhibition of 5-HT synthesis by chronic

parachlorophenylalanine treatment (Manrique et al., 1994; Compan et al., 1998). Thus the

5-HT1B receptor up-regulation after cocaine withdrawal may be a consequence of

impaired 5-HT neurotransmission.

- Effects on the amygdala

In the amygdala, saline-treated females presented a decrease in the levels of DA

and DOPAC from 5 to 10 days after the last administration, with this effect not being

observed in cocaine-treated females, which suggests an effect of cocaine treatment on

the evolution pattern of the dopaminergic neurotransmission in the amygdala of female

rats during long-term withdrawal. In fact, although no significant differences were

observed in DOPAC/DA turnover rate, 10 days after withdrawal cocaine-treated females

presented higher levels of DOPAC as compared to control females. Moreover, at this

time, saline-treated females presented lower levels of DOPAC as compared to saline-

treated males. This sex difference was not present in the cocaine group, and it is likely to

result from an altered DA system maturation in the amygdala of cocaine-treated female

rats. The expression levels of D1, D2 and D3 receptors, DAT and MAO-A mRNA in male

rats, determined 10 days after withdrawal also did not showed any treatment effects.

Altogether these data suggest a sex-effect regarding dopaminergic neurotransmission in

the amygdala during long-term withdrawal, with females being more sensitive than do

males to the effects of cocaine. Amygdala continues to mature during adolescence (Cao

et al., 2007), and therefore the alterations observed could be due to the cocaine effects on

the developmental process occurring during the exposure period.

Withdrawal from cocaine use is accompanied by the development of negative

emotional states characterized by intense dysphoria and anxiety (Koob and Le Moal,

267

1997; Gordon and Rosen, 1999). These observations suggest a role for the amygdala, a

component of the limbic system that serves an essential function in associative emotional

learning (Miserendino et al., 1990; Fanselow and LeDoux, 1999; LeDoux, 2000; Nader et

al., 2000; Maren, 2001). The amygdala receives dopaminergic projections from the VTA

(Koob et al., 1998; Nestler, 2001), that has been implicated, along with other brain

regions, in drug-seeking behaviour and relapse both in humans and experimental animals

(Breiter et al., 1997; Kalivas and McFarland, 2003). Because the amygdala sends

projections to key components of reward circuitry, including the Nac and prefrontal cortex

(Baxter and Murray, 2002; Wise, 2002), synaptic modifications in the amygdala resulting

from cocaine use may affect the function of these reward-associated brain structures.

Alternatively, the amygdala may contribute to brain reward function through the formation

of associations between the rewarding effects of cocaine and contextual cues (Hayes and

Gardner, 2004).

On the other hand, although serotonergic neurotransmission in the amygdala is

associated with anxiety-like behaviour (Higgins et al., 1991; Gargiulo et al., 1996;

Zangrossi et al., 1999), and anxiety has been descried as a symptom of cocaine

withdrawal (Gawin, 1991), our data seems to indicate that cocaine treatment throughout

adolescence did not have long-term effects in basal serotonergic neurotransmission in the

amygdala, since there were no alterations in the levels of 5-HT and 5-HIAA or in 5-HT

turnover rate, both 5 and 10 days after withdrawal, for both sexes.

- Effects on the hypothalamus

In the hypothalamus, cocaine treatment did not affect the levels of DA, 5-HT, their

metabolites, or turnover rates, neither in males or females as assessed 5 and 10 days

after withdrawal. Moreover, the expression levels of D1, D2 and D3 receptors, DAT and

MAO-A mRNAs were also not altered 10 days after withdrawal in cocaine-exposed males.

These data obtained in the hypothalamus seem to complement the hormonal data

indicating that chronic cocaine binge administration does not seem to have prolonged

effects upon the HPA axis activity under basal conditions, both in male and female rats.

Nevertheless, other authors have reported alterations in dopaminergic and

serotonergic neurotransmission in the hypothalamus during cocaine withdrawal. Karoun

and colleagues (2004) reported that, 1 week after 7 days of repeated cocaine

administration, the DA turnover in the hypothalamus was reduced (Karoum et al., 1994).

Differences in treatment duration, doses and period of exposure (adulthood versus

268

adolescence) may be underlying the differences between our study and the results

reported by the latter study.

- Effects on the dorsal raphe nucleus

Since symptoms of depression have been associated with cocaine withdrawal, and

a dysfunction of the serotonergic neurotransmission is thought to underlie a variety of

psychiatric disorders, such as depression (Arango et al., 2002; Lira et al., 2003; Bach-

Mizrachi et al., 2006), it was expected that serotonergic neurotransmission in the dorsal

raphe nucleus would be altered during withdrawal from chronic binge cocaine. However,

in dorsal raphe nucleus, no effects of chronic binge cocaine exposure were observed on

the levels of 5-HT and 5-HIAA or 5-HT turnover rate, both in male and female rats, as

assessed 5 and 10 days after cocaine withdrawal. Nevertheless, 5 days after the last

administration, saline-treated males presented higher levels of DA as compared to saline

females, with this sex effect not being observed in the cocaine-treated group, which may

indicate some disruption of the dopaminergic neurotransmission in the dorsal raphe

nucleus. It is known that 5-HT neurons of the dorsal raphe nucleus receives a dense

innervation from midbrain dopaminergic neurons (Peyron et al., 1995; Kitahama et al.,

2000) and express D2 and D3 receptors (Mansour et al., 1990; Suzuki et al., 1998).

These anatomical evidences suggest that DA input to the dorsal raphe nucleus may play a

critical role in the regulation of the dorsal raphe nucleus 5-HT neurons activity.

Consistently with this notion, in vivo neurochemical studies have reported that

administration of DA receptor agonists increases the synthesis and release of 5-HT in the

dorsal raphe nucleus (Ferre et al., 1994; Matsumoto et al., 1996). Direct evidence for a

role of DA in the regulation of 5-HT neurons activity comes from an in vitro

electrophysiological study showing that activation of DA receptors induces a membrane

depolarization and thereby increases the excitability of dorsal raphe 5-HT neurons (Haj-

Dahmane, 2001). However, the alteration on dopaminergic neurotransmission observed 5

days after withdrawal was not accompanied by any alteration on the levels of 5-HT, 5-

HIAA or 5-HT turnover rate. Moreover the altered dopaminergic activity was no longer

present 10 days after withdrawal.

- Effects on the cerebellum

The long-term effects of exposure to psychostimulants on 5-HT neurotransmission

in the cerebellum are not well characterized. Our data indicate that the chronic binge

cocaine administration throughout adolescence did not have long-term effects on the

269

levels of 5-HT, 5-HIAA or 5-HT turnover rate. These results are consistent with those

obtained in the dorsal raphe nucleus, and altogether seem to point to an absence of long-

term effects of chronic cocaine administration throughout adolescence. Nevertheless,

saline-treated males presented decreased levels of HVA from 5 to 10 days after the last

administration, but no differences to cocaine-treated males were observed at 5 and 10

days of withdrawal, suggesting an effect of cocaine on the evolution pattern of the

dopaminergic neurotransmission, from 5 to 10 days after withdrawal.

- Interaction between the dopaminergic system and the HPA axis

As already stated in chapter 1, the interaction of glucocorticoids with the

mesocorticolimbic DA system, through the GR, may have a significant impact on the

vulnerability to self-administer psychostimulant drugs, since increased dopaminergic

transmission in these pathways is critical for the reinforcing properties of abusive drugs,

rendering the individuals more susceptible to drug reinforcement (Roberts et al., 1980;

Koob and Le Moal, 1997; Marinelli and Piazza, 2002). However, our results seem to

suggest that the increased dopaminergic activity observed 10 days after withdrawal in

cocaine-treated male rats was not associated with an augmented stimulatory effect of the

HPA axis, since no effects of cocaine were observed in basal corticosterone levels, and

the expression levels of GR mRNA was only affected in the SN-VTA of cocaine-treated

males that presented lower expression levels as compared to control animals at 10 days,

suggesting that this was not the mechanism involved in the increased dopaminergic

activity observed.

- Conclusion

In conclusion, chronic exposure to cocaine throughout the adolescence produced

long-lasting changes upon the dopaminergic system in male rats, with the data obtained

suggesting an increase in dopaminergic activity of the nigrostriatal pathway. In fact, the

data pointing to increased dopaminergic activity, is supported by an increase in the

expression levels of TH mRNA found in the area of origin of the dopaminergic system.

The increased dopaminergic activity may represent an attempt to compensate for a

chronic cocaine-induced depression of the dopaminergic system. The serotonergic activity

in male rats was also long-term affected by cocaine treatment, Increased 5-HT levels in

SN-VTA were observed, and just like as suggested for the dopaminergic activity, the

increase in the serotonergic activity may represent an attempt to counteract the

downregulation of this system’s activity during withdrawal.

270

Diverging from the data obtained in male rats, in cocaine-treated females it was

observed an increase of the dopaminergic activity in the amygdala and in the

hippocampus, as well as an increase of the serotonergic activity in the hippocampus and

in the Nac throughout long-term withdrawal, these neurochemical alterations, among other

effects, could contribute to an increased vulnerability to relapse and interfere with

processes of learning and memory formation.

4.4.4. Effects of chronic cocaine treatment during adolescence on the anxiogenic-like behaviour

As already discussed in chapter 3, symptoms of anxiety and depression are

associated with withdrawal from cocaine exposure, particularly in the initial period

(Gawin, 1991). It has been postulated that anxiety persists into the later withdrawal period

and is accompanied by intense craving that may lead to relapse.

The number of entries and the time spent in the aversive open arms of the EPM

are typical measures of anxiety in this behavioural test (Pellow et al., 1985). We did not

observed both 5 and 10 days after cocaine withdrawal effects on these measures in male

rats. The locomotor activity was also not affected by the treatment, both 5 and 10 days

after withdrawal, as indicated by the absence of differences between cocaine- and saline-

treated males, in the number of closed arm entries, the most reliable measure of

locomotor activity (Hogg, 1996).

Nevertheless, data analysis indicate that male rats exposed to cocaine during

adolescence presented 10 days after withdrawal, a decrease in the time spent in the

central platform. The time spent in the center portion of the EPM has been interpreted as

a decisional component of approach/avoidance conflict task (Rodgers and Johnson, 1995;

Carobrez and Bertoglio, 2005). These results add to the alterations in risk-assessment in

young adult male rats pre-exposed to cocaine during adolescence, when exploring the

EPM apparatus.

There are numerous studies reporting that animals exhibit enhanced anxiety

during the first days after cocaine withdrawal (Sarnyai et al., 1995; Basso et al., 1999).

However, the extent to which anxiety-like responses persist over extended drug-free

periods is unclear. In some studies, cocaine pre-exposed animals exhibited anxiety-like

behaviours after drug-free periods of up to 7 days (e.g., Fontana and Commissaris, 1989;

Gordon and Rosen, 1999), whereas in other studies, cocaine pre-exposure was

ineffective in elevating anxiety beyond the first 2–3 days of withdrawal (Mutschler and

271

Miczek, 1998). Erb and colleagues (2006) reported enhanced levels of anxiety in adult

rats, 10 days after cocaine withdrawal (30 mg/kg, i.p. for 7 days), but only when animals

were exposed to an environment previously paired with cocaine, suggesting that a

“reminder” of the drug experience, such as re-exposure to contextual cues previously

paired with cocaine, may be necessary to reactivate a cocaine-induced anxiety-like state

after the initial withdrawal period (Erb et al., 2006).

To our knowledge, only one study, by Estelles and colleagues have been

performed to evaluate the later impact of cocaine exposure during adolescence on anxiety

like-behaviour (Estelles et al., 2007). In mice, a binge pattern of cocaine administration

during adolescence modified the anxiety of the treated-adolescent, increasing the time

spent on the open arms of the EPM; however, housing in isolation counteracted this

cocaine effect (Estelles et al., 2007). In fact, it has been suggested that housing condition

can modulate the behavioural responses of rats to the EPM (Starkey et al., 2007). In any

animal model, the utility of the EPM is dependent on the anxiety threshold levels, low

anxiety baselines are less than optimal for detecting anti-anxiety effects and high

baselines virtually useless for detecting anxiety enhancement (Rodgers and Cole, 1993).

For example, six weeks of isolation reared rats spent less time into the open arms on

EPM compared to group housed rats (Gupta and Rana, 2007). Thus, in the present study,

the fact that animals were housed individually may have contributed to a high anxiety

baseline, which mask eventual effects of cocaine.

As already discussed in this chapter, 10 days after cocaine withdrawal, the basal

levels of corticosterone were not significantly altered in male rats, while, however after the

EPM, cocaine-treated males presented a significant increase in corticosterone, suggesting

that although chronic cocaine administration during adolescence does not seem to have

prolonged effects upon corticosterone levels under basal conditions, it does sensitize the

HPA axis activity under anxiogenic conditions.

One of the most consistently described biological abnormalities in depressive and

anxiety disorders is dysregulation of the HPA axis, which is typically normalized by

successful antidepressant or anxiolytic therapy (Nestler et al., 2002; Wichniak et al., 2004;

Mikkelsen et al., 2005). However, our data showed that the HPA axis activity was long-

termed affected, but this was not accompanied by alterations in anxiety-like behaviour as

observed in the EPM. Munoz-Abellan et al. (2008) have already reported dissociation

between HPA axis activation and the anxiety-like behaviour observed during EPM. These

authors suggested that after a very short exposure to the EPM, HPA axis activation may

reflect arousal rather than a degree of emotional reaction to the situation (Munoz-Abellan

et al., 2008).

272

Corticosterone, either stress-induced or exogenously administered, is clearly

implicated in anxiety in the EPM in adult rats (e.g., Adamec et al., 2006; Matuszewich et

al., 2007). For example, corticosterone administered to the amygdala increased anxiety-

like behaviour in the EPM (Shepard et al., 2000; Myers et al., 2005) and HPA axis

response to stress (Shepard et al., 2003). Exposure to an acute stressor before testing in

the EPM increased anxiety-like behaviour, which could be attenuated by blockade of

corticosterone synthesis and reinstated by administering corticosteroid agonists (Calvo

and Volosin, 2001). Moreover, antagonists of the corticosteroid receptors attenuated the

anxiogenic effects of pre-exposure to acute stress in the EPM (Korte et al., 1995).

However, in non-stressed rats, individual differences in corticosterone concentrations

while in the EPM showed a stronger association with risk-assessment than either with

anxiety (open arm exploration) or locomotor activity (Rodgers et al., 1999; Albrechet-

Souza et al., 2007). Based in the aforementioned, the higher levels of corticosterone

displayed by cocaine exposed males after the EPM, at 10 days of withdrawal are most

likely related with the impaired risk-assessment observed in these animals, as supported

by the decreased time spent in the central platform of the maze.

The molecular mechanisms by which pretreatment with cocaine throughout

adolescence lead to sensitization of the HPA axis to an anxiogenic stimulus were not

addressed by the present study. One possible explanation to the augmented HPA axis

response to the EPM in the cocaine exposed males could be related with an impaired GR-

mediated negative feedback, which would remove inhibitory constraint from the HPA axis,

thus augmenting the glucocorticoid response to the EPM. However, and as already

mentioned in this chapter, 10 days after withdrawal, no treatment effects were found in the

expression levels of the GR mRNA in pituitary and in several brain areas implicated in

negative feedback regulation of the HPA axis. Although these findings suggest that the

augmented HPA axis response was independent of impaired GR-mediated feedback

inhibition, functional measures of feedback (e.g., sensitivity to dexamethasone

suppression) are necessary to clarify the contribution of altered negative feedback.

Further investigation is required to determine the role of adaptations within the HPA axis

that are independent of GR-mediated feedback in the intensified response.

The neurochemical results obtained after the EPM, 10 days after withdrawal,

showed that the dopaminergic and the serotonergic response in amygdala of cocaine-

treated male rats were affected under an anxiogenic situation, as indicated by the lower

levels of DOPAC and 5-HIAA, as compared to control males. In fact, control males display

an increase in DOPAC levels after the EPM that was not observed in cocaine-treated

males. As already discussed in Chapter 3, amygdala is a critical structure in the neural

273

pathways necessary for implementing advantageous decisions. The amygdala is involved

in the attachment of emotional valence to events (i.e., reward and punishment) (Bechara

et al., 1999) and is crucial for decision making and learning (e.g., Winstanley et al., 2004).

Lesions of the amygdala decrease harm avoidance (Johansson and Hansen, 2002). Thus,

the disruption in neurotransmission observed in the amygdala of cocaine-treated males

after the EPM may be related with the impaired risk assessment observed in these

animals, as suggested by the decreased time spent in the central platform of the maze.

and by this way contributing to a increased vulnerability to relapse.

These neurochemical results differ substantially from the data obtained in the

EPM 2 days after withdrawal (discussed in Chapter 3). During acute withdrawal, a

significant altered prefrontal cortex neurotransmission was observed in cocaine-treated

male rats, that was no longer found after long-term withdrawal. Besides the changes

observed in amygdala, no significant differences were observed in the neurotransmitter

profile between cocaine-treated and controls in other brain areas. However, when

comparing the profiles before and after EPM were observed important differences

between cocaine- and saline-treated male rats.

In the prefrontal cortex cocaine-treated males after EPM presented higher

DOPAC/DA turnover rate as compared to cocaine males under basal conditions and this

effect was not observed in saline-treated males, which represents a decreased availability

of DA in this region. In the hippocampus, control males presented higher 5-HT turnover

rate after EPM as compared to basal conditions, however this effect was not observed in

cocaine-treated males, which may represent an altered use of 5-HT.

In 2005, Carvalho and colleagues reported the effects of EPM exposure in the

biogenic amine content of the prefrontal cortex, amygdala, dorsal hippocampus of rats

(Carvalho et al., 2005). Their results suggest that the exposure to EPM causes a

significant decrease in the 5-HT concentration in the prefrontal cortex, amygdala and

dorsal hippocampus; with the amygdala being the only structure in which there was also a

reduction in the tissue concentration of DA and DOPAC. Moreover, their results suggest

that the observed changes in the neurotransmission of 5-HT and DA could not be

attributed to enhanced metabolism, since the turnover rates of these biogenic amines

were not different from those of control animals, not tested in the EPM. These results are

quite different from those obtained in our study. Several causes may underlie these

differences. One possible explanation may be the previous experimental manipulation that

animals in our study were submitted to, including the repeated injection protocol and long-

term social isolation, since both procedures are known to be stressful and capable of

274

interfere with neurochemical processes (Suthanthirarajan and Subrahmanyam, 1983;

Persico et al., 1995; Kosten et al., 2003), this way affecting the response to the EPM.

In the SN-VTA, after the EPM, no cocaine treatment effects were detected in the

male rats. Nevertheless the higher levels of DOPAC observed in cocaine-treated males as

compared to controls under basal conditions was not found in the animals that performed

the EPM.

In the dorsal raphe nucleus, after the EPM, in both cocaine- and saline-treated

males the levels of 5-HT were higher after the EPM as compared to basal levels. The

levels of 5-HT in dorsal raphe nucleus are related with anxiety-like behaviour.

Microinfusions of drugs that increase serotonergic activity directly into the dorsal raphe

nucleus, like the benzodiazepine receptor partial inverse agonists N-methyl-beta-

carboline-3-carboxamide (FG-7142) (Lista et al., 1990), methyl 6,7-dimethoxy-4-ethyl-

beta-carboline-3-carboxylate (DMCM) (Maier et al., 1995), and the anxiety-related

neuropeptide urocortin 2 (Hammack et al., 2003; Amat et al., 2004) increase anxiety

related behavioural responses. In contrast, microinfusions of 5-HT or 5-HT1A agonists

into the dorsal raphe decrease anxiety-related behavioural responses, presumably via

activation of somatodendritic 5-HT1A autoreceptors on serotonergic neurons, resulting in

decreased 5-HT neurotransmission (Higgins et al., 1988; Higgins et al., 1992; Schreiber

and De Vry, 1993). In the present study the 5-HT turnover rate was lower in saline-treated

males after the EPM as compared to basal levels, suggesting a decrease in the 5-HT

metabolism. This effect was not observed in cocaine-treated males, which could be a sign

of increased anxiety-like behaviour, and account to explain the different strategies to

explore the EPM apparatus, as indicated by the decreased time spent in the central

platform by the cocaine-treated male rats.

Interestingly the higher levels of corticosterone were not accompanied by altered

DA or 5-HT neurotransmission in the hypothalamus. Nevertheless, a mechanism of

cocaine-induced supersensitivity of 5-HT2A receptors, in the paraventricular nucleus, have

been described after 42-h withdrawal from repeated cocaine treatment (15 mg/kg i.p.,

twice a day for 7 days) (Levy et al., 1992b). Activation of 5-HT2A receptors in the

hypothalamic paraventricular nucleus increases the secretion of ACTH, corticosterone,

oxytocin, and prolactin (Van de Kar et al., 2001). Thus, a possible alteration in the 5-HT

receptors can not be excluded.

To summarize, in the present study it was demonstrated that the EPM-induced

activation of the HPA axis is augmented in prolonged cocaine withdrawal and that despite

the absence of increased anxiogenic-like behaviour, the impaired risk-assessment

275

observed in cocaine-treated it is likely to be related with the altered HPA axis response.

Moreover, the neurotransmission disruption observed in amygdala of cocaine-treated

males in response to the EPM may also be related with changes in the behavioural

strategy adopted to cope with the anxiogenic stimulus provided by the EPM. Altogether,

these results may point to an increased susceptibility to drug use after long-term

withdrawal, since impaired risk-assessment and enhanced HPA axis response to an

environmental challenge are considered risk factors for relapse. Stress-induced HPA axis

responses can be used to predict the likelihood of drug relapse (Sinha et al., 2006), and

risk-taking behaviours are associated with increased risk for drug use onset (Rios-Bedoya

et al., 2008). Therefore, the present results may also contribute to increase knowledge on

how to structure drug abuse prevention.

4.4.5. Effects of chronic cocaine treatment during adolescence on the aggressive behaviour

In a variety of vertebrates, including primates, correlations have been established

between levels of aggressiveness and testosterone or neural testosterone metabolism

(Rose et al., 1971; Barfield et al., 1972; Albert et al., 1986). In mice, strong correlations

have been established between testosterone and the latency to attack an intruder in

resident-intruder models of offensive aggression (Van Oortmerssen et al., 1987). In rats,

castration results in a decline of aggression (Albert et al., 1986). Presumably, testosterone

would affect neural networks controlling offensive aggression, resulting in an enhanced

arousal and predisposition towards agonistic displays (Delville et al., 1996). Indeed,

testosterone is suspected to modulate offensive aggression by affecting several areas

within the limbic system (Bean and Conner, 1978; Albert et al., 1987).

As already discussed in this chapter, the present study showed that 10 days after

cocaine withdrawal male rats presented decreased levels of testosterone. Thus, due to

the role of testosterone in the aggressive behaviour, it was hypothesized that aggressive

behaviour in these animals could be altered.

There are several reports that have found an increase in aggressive and violent

behaviours in cocaine addicts (Denison et al., 1997; Chermack and Blow, 2002; Fals-

Stewart et al., 2003; Moore et al., 2008). The preclinical literature linking cocaine and

aggression is not so cohesive probably because a variety of test paradigms, animal

models, and dosing regimens have been utilized, yielding conflicting data (Miczek, 1979;

Hadfield et al., 1982; Emley and Hutchinson, 1983; Filibeck et al., 1988; Darmani et al.,

1990; Long et al., 1996). For example, Mizcek (1979) reported an inverse relationship

276

between cocaine treatment and aggression in mice treated acutely with medium- and

high-dose (8.0 and 32.0 mg/kg, respectively) cocaine, while reporting no effect on

aggression at low doses (0.5 and 2.0 mg/kg). In comparison, adult male rats treated

chronically with low doses of cocaine (1.0 mg/kg) showed elevated measures of

aggression (Long et al., 1996).

A well-established and ethologically valid model of offensive aggression used

routinely to test for aggressive behaviour is the resident–intruder paradigm (Miczek, 1979;

Koolhaas et al., 1980). In the present study, this paradigm was used to examine the long-

term effects of chronic cocaine exposure during adolescence on aggressive behaviour of

young adult male rats. The behavioural data obtained revealed no long-term effects of

chronic cocaine administration throughout adolescence on the aggressive behaviour of

the male rats, when assessed 10 days after withdrawal.

There are fewer studies evaluating the long-term effects of cocaine on social

behaviours. An increase in aggression has been found in adult rats exposed gestationally

to cocaine, using the resident-intruder paradigm (Johns et al., 1994) or toward a rival

when competing for water (Wood and Spear, 1998). Although our and the later mentioned

studies present data from long-term effects of cocaine exposure, the results can not be

compared, since the time of cocaine exposure were different (adolescence versus

gestational development). In a study using adolescent hamsters, Harrison and colleagues

(2000) also reported that chronic cocaine treatment increased the aggressive behaviour

(Harrison et al., 2000); however, this data was obtained 24 hours after the last drug

administration and in the present study aggressive behaviour was assessed 10 days after

the last drug administration. Nevertheless, in mice, Estelles and colleagues (2005)

reported that animals prenatally treated with cocaine did not exhibit an increase in

aggressive behaviours when tested as adults (Estelles et al., 2005).

In studies evaluating social behaviour, the social history of the animals employed

is very important. Isolated mice exhibit a particular behavioural profile with higher levels of

aggression than those housed in groups (White et al., 1991; Matsumoto et al., 2005). In

male rats social isolation is also reported to increase aggression (Wongwitdecha and

Marsden, 1996; Hall, 1998). Thus, the social isolation that animals in the present study

were submitted to could have increased the aggressive behaviour, by this way masking

the cocaine effects regarding this behaviour.

Of particular interest, however, was the finding that cocaine-treated male rats

displayed increased duration of the anogenital sniffing behaviour. Moreover, these

animals also exhibited lower frequency of and increased latency to flight behaviour. These

277

results appear to indicate that cocaine treatment during adolescence led to an increase in

social investigation and reduce fear at 10 days after withdrawal. This seems to contradict

the established consensus regarding the reduction in social contacts within conspecifics

that has been observed after cocaine administration in rodents (Darmani et al., 1990;

Rademacher et al., 2002). However, this assumption seems not to be so linear, as for

instance Estelles at al. (2005) reported that, isolated mice show a significant decrease in

the time dedicated to social interactions, while grouped animals prenatally treated with

cocaine exhibited an increase in the social contact and the time dedicated to each

contact, showing that some variables can influence the cocaine effects on social

investigation. In adult rats social isolation is reported to increase social investigation of an

unfamiliar rat (Niesink and van Ree, 1982; Varlinskaya et al., 1999). Therefore, rats are

routinely isolated prior to being tested in the social interaction paradigm, as this should

increase the time spent interacting with conspecifics, making alterations at this level

easier to observe (File and Seth, 2003). In our study, and contrarily to that reported by

Estelles and colleagues (2005), cocaine treatment seems to have intensified the effects of

social isolation in what concerns to the social investigation. The species in study (rat

versus mice), the time of exposure to the drug (adolescence versus gestational

development) and the withdrawal status (10 days after the last administration versus since

birth to adulthood) may have contributed to the different results between these studies.

Behavioural data suggest that chronic cocaine exposure during adolescence have

long-lasting influences in male rat reactions to social threat, providing some indications to

the capability of cocaine during adolescence to affect the ability of young adult rats to

interact properly with conspecifics. Moreover, the reduced fear presented by the cocaine-

treated animals, as indicated by the decrease in flight behaviour, suggest an altered risk-

assessment, supporting the results obtained in the EPM, also indicating an effects of

cocaine treatment in the risk-assessment.

In this study the corticosterone response to the resident-intruder paradigm was

also determined in order to evaluate the effects of cocaine treatment in the HPA axis

response to a social threat. HPA axis abnormalities in humans and rodents have often

been associated with changes in male aggression (McBurnett et al., 2000; de Kloet, 2003;

Haller et al., 2004). Paradoxically, high as well as low HPA axis activity can be associated

with excessive aggression (reviewed by Veenema and Neumann, 2007). High HPA axis

activity, often associated with a state of hyperarousal, is thought to underlie sudden

outbursts of aggression, whereas chronically low HPA axis activity, often associated with

a state of hypoarousal, might induce long-lasting changes in brain functions that promote

violence (Haller and Kruk, 2006). In clinical studies, elevated glucocorticoid responses

278

were found in individuals with high aggression levels (Gerra et al., 1997). Furthermore, an

acute activation of the HPA axis was found to promote aggressive behaviour in rodents

(Kruk et al., 2004; Mikics et al., 2004). In contrast, there are preclinical (Haller et al., 2001;

Haller et al., 2004) and clinical studies (McBurnett and Lahey, 1994; McBurnett et al.,

2000; Shoal et al., 2003) reporting an inverse relationship between pituitary-adrenocortical

(re)activity and aggressive behaviour. However, in these studies, high levels and

abnormal forms of aggression were associated with a chronic glucocorticoid deficiency.

The levels of corticosterone after the resident-intruder paradigm, although higher

than the basal, did not differ between cocaine- and saline-treated males, suggesting that

cocaine treatment did not affect the HPA axis response to the social threat provided by

the resident-intruder paradigm. As this behavioural test is known to induce a very large

adrenocortical stress response both in resident and intruder rats (Schuurman, 1980), the

increased response of the HPA axis induced by the test was the expected one. However,

it is also possible that this way mask the effects of cocaine treatment.

Classical neuroanatomical tracing and electrochemical lesion/stimulation studies

have revealed the global neural substrates of aggression (Luiten et al., 1985; Gregg and

Siegel, 2001). Several regions of cortex, amygdala, septum, hypothalamus,

periaqueductal gray and their interconnected structures are among the best documented

and characterized in this respect. In addition, more recent studies using functional

immediate-early gene expression mapping (i.e., c-FOS, p-CREB, zif-268) start to yield a

more detailed picture of the individual neurons and their neurochemical identities that

become activated within these brain regions during the expression of aggressive

behaviour (Gammie and Nelson, 2001; Halasz et al., 2002; van der Vegt et al., 2003b;

Veening et al., 2005). Despite this expansion, the 5-HT neurotransmitter system remains

the primary molecular determinant of aggression (Tuinier et al., 1995; Berman et al., 1997;

Miczek et al., 2002).

There exists a large body of evidence implicating the 5-HT as regulator of

aggressive behaviour in a number of animal models of aggression (Hadfield et al., 1982;

Vergnes et al., 1988; Higley et al., 1992; Kyes et al., 1995) and in adolescent and adult

human populations (Brown et al., 1982; Linnoila et al., 1983; Kruesi et al., 1990). In

rodents, aggressive behaviour is effectively reduced by treatment with 5-HT1A and 5-

HT1B receptor agonists (De Almeida and Lucion, 1997; Miczek et al., 1998; Simon et al.,

1998; de Boer et al., 1999; Ferris et al., 1999). In humans, excessive aggression and

impulsive violent behaviours are associated with low cerebrospinal fluid levels of the 5-HT

metabolite 5-HIAA (Berman et al., 1997). In fact, the prevailing view has been that 5-HT

exerts an inhibitory control over aggression (Linnoila and Virkkunen, 1992; Kavoussi et al.,

279

1997). However, other findings in animal research challenge this 5-HT deficiency

hypothesis of aggression, in which 5-HT is positively associated with the display of

aggression (Olivier, 2004; de Boer and Koolhaas, 2005). These opposing views might

demonstrate important differences in 5-HT functioning in abnormal or pathological forms

of aggression compared to normal and adaptive levels of aggression. Likewise, there may

be essential differences regarding the role of the 5-HT system in trait-like and state-like

aggression. Trait-like aggression can be associated or induced by chronically reduced 5-

HT activity (Miczek et al., 2002), whereas state-like aggression is characterized by

increased 5-HT activity (van der Vegt et al., 2003a; van der Vegt et al., 2003b; Summers

et al., 2005). It has also been demonstrated that 5-HT plays an important role in cocaine-

mediated aggressive behaviour (Moeller et al., 1994; DeLeon et al., 2002; Ricci et al.,

2004).

In the present study, the absence of differences in aggressive behaviour between

cocaine- and saline-treated animals was followed by the absence of differences in the

levels of 5-HT or 5-HIAA in the dorsal raphe nucleus after the resident-intruder paradigm.

Moreover, both in control and cocaine-treated rats, the basal levels of 5-HT and 5-HIAA

also did not differ from the levels after the resident-intruder paradigm. However, in saline-

treated rats the 5-HT turnover rate decreased between basal and post-test conditions, and

this effect was not observed in cocaine-treated rats. This may point to an effect of cocaine

treatment in the 5-HT neurotransmission in the dorsal raphe nucleus of rats facing the

social conflict provided by the resident-intruder paradigm.

Moreover, in the hippocampus, the levels of 5-HT were increased in the saline-

treated rats after the resident-intruder paradigm, and again this effect was not observed in

cocaine-treated males. Adding to this in the amygdala the 5-HT turnover was decreased

after the test only in the cocaine-treated males.

The serotonergic system modulates aggressive behaviour in interaction with other

neurotransmitters, of which the corticolimbic DA continues to be of interest for its critical

role in integrating motivation and motor functions (Robbins et al., 1989).

In the present study, the levels of 5-HT after the resident-intruder paradigm in the

prefrontal cortex did not differ between cocaine- and saline-treated males, as well as no

significant differences were observed in the 5-HT neurotransmission in the prefrontal

cortex between basal and post-test conditions. Damage to or pharmacological inhibition of

the prefrontal cortex can increase aggression, and this effect is hypothesized to be

caused by loss of impulse control (Tobin and Logue, 1994). In a former microdialysis

study, decreases in 5-HT and increases in DA levels in prefrontal cortex were observed

280

using the resident-intruder paradigm (van Erp and Miczek, 2000). Differences in the

experimental procedure may be responsible for the discrepancy of data obtained in the

present study and the data reported by Van Erp and Miczek (2000), namely the housing

condition (male rats housed individually versus male rats housed with a female) since it is

known that social isolation induce an alteration of the serotonergic system in rats (Rilke et

al., 1998; Dalley et al., 2002; Bibancos et al., 2007). Nevertheless, the behavioural test

induced a decrease in the levels of HVA in cocaine male rats. In fact, after the test these

animals presented a decrease in the DA metabolism as indicated by the decreased

HVA/DA turnover rate. Moreover, the dopaminergic neurotransmission was also affected

in hippocampus, both by the test and by the cocaine treatment. In the hippocampus, the

aggressive test induced an increase in DA levels in control animals that was not observed

in the cocaine-treated animals, suggesting a cocaine effect in the dopaminergic response

to the behavioural test. Indeed, after the test the levels of DOPAC were lower in cocaine-

treated animals as compared to control animals. These data suggest a decrease in the

dopaminergic neurotransmission in cocaine-treated rat in response to the resident-intruder

paradigm as compared to control animals.

In the amygdala, the levels of HVA after the test were reduced to non detectable

levels in both cocaine- and saline-treated animals, suggesting an effect of the test on the

dopaminergic neurotransmission in the amygdala.

In the SN-VTA, the dopaminergic response to the behavioural test was affected by

the cocaine treatment, as the levels of DOPAC in the cocaine-treated rats were reduced

by the test. Also in SN-VTA the HVA levels became undetectable after the test, both in

cocaine- and saline-treated rats. In this area the serotonergic neurotransmission was also

affected by the treatment, as after the test the 5-HT metabolism was lower in cocaine-

treated rats as compared to control animals. Moreover, in cocaine-treated rats the levels

of 5-HIAA were decreased by the test and this effect was not observed in control animals.

In the present study the serotonergic response in the hypothalamus to the social

conflict provided by the resident-intruder paradigm was affected by the cocaine treatment,

as indicated by the decreased 5-HT metabolism in the cocaine-treated rats that was not

observed in the control animals. In this area it was also observed an effect of the test on

the DA neurotransmission, as the behavioural test induced a decrease in the

dopaminergic metabolism of both cocaine- and saline-treated rats, suggested by the

decreased DOPAC/DA turnover rate. Also in this area the HVA levels become

undetectable in both cocaine and control animals after the test. Nevertheless, the DOPAC

levels were decreased by the test only in the cocaine-treated rats, suggesting a potential

effect of the cocaine treatment in the dopaminergic response to the social conflict

281

provided by the resident-intruder paradigm. It seems like cocaine treatment decreased

both serotonergic and dopaminergic response of hypothalamus to the social stressful

situation provided by the resident-intruder paradigm, but these alterations were not

reflected in the HPA axis activity, since no differences were observed between cocaine-

and saline-treated animals regarding the plasma levels of corticosterone.

Taken together, these findings indicate a decrease in the dopaminergic activity in

response to the social conflict provided by the resident-intruder paradigm in the cocaine-

treated rats, 10 days after withdrawal. Cocaine treatment also seems to have influenced

the serotonergic response to the challenge provided by the test, as data obtained in this

study indicates an increased serotonergic activity in the dorsal raphe nucleus and in the

hippocampus, while decreased in the amygdala, prefrontal cortex, hypothalamus and SN-

VTA. However, these alterations in the dopoaminergic and serotonergic

neurotransmission were not reflected on alterations in the aggressive behaviour. Rather,

these data may be relevant to explain the increased social investigation and decreased

defensive behaviour observed in cocaine-treated rats. Namely, the alterations in the

amygdala and prefrontal cortex were probably related with the decreased defensive

behaviour observed in cocaine-treated male rats. The decreased defensive behaviour

appears to be related with a decrease in fear, which may result from an impaired risk

assessment, also observed in these animals when exploring the EPM, and as was already

discussed, altered function in these brain regions are associated with increased risky

behaviour (Bechara, 2001; Blanchard et al., 2005; Muller and Fendt, 2006). On the other

hand, the knowledge of the hippocampus key role in the individual recognition

(i.e.Terranova et al., 1994; Kogan et al., 2000), lead us to suggest that the increase in

social investigation observed in cocaine-treated male rats, that was probably related with

an impaired ability to learn and remember individual (see, Yamamuro, 2006), was mostly

caused by the altered function of the hippocampus.

4.5. Conclusion

The data obtained in this study showed that cocaine exposure throughout

adolescence induced sex-dependent long-lasting hormonal, neurochemical and

behavioural effects.

Chronic cocaine exposure throughout adolescence led to long-lasting disruptive

effects on the HPG function in male rats, as indicated by the decrease in testosterone

levels observed 10 days after cocaine withdrawal, with these animals also showing a

decrease in relative gonadal weight.

282

Chronic cocaine exposure did produce long-lasting changes upon the

dopaminergic system in male rats. The neurochemical data obtained in the present study

showed an increase of the dopaminergic activity in the nigrostriatal pathway, also

supported by the increased expression levels of TH mRNA in the SN-VTA. This process

may represent an attempt to compensate for a chronic cocaine-induced depression of the

dopaminergic system.

Long-lasting effects of cocaine treatment were also found in the serotonergic

activity of the male rats. Increased 5-HT levels in SN-VTA were observed in cocaine-

treated males, and as suggested for the dopaminergic activity, such increased

serotonergic activity may represent an attempt to counteract the downregulation of this

system during withdrawal.

Diverging from the data obtained in male rats, cocaine-treated females presented

an increase in the dopaminergic activity in the amygdala and hippocampus, and an

increase in serotonergic activity in the hippocampus and Nac throughout long-term

withdrawal. Among other effects, these long-term alterations could contribute to an

increased vulnerability to relapse and interfere with processes of learning and memory

formation.

After prolonged withdrawal, impaired risk-assessment was observed in cocaine-

treated male rats when exploring the EPM at 10 days after withdrawal. Simultaneously,

disrupted neurotransmission in the amygdala was observed in these animals in response

to the EPM, which may be related with changes in the behavioural strategy adopted to

cope with the anxiogenic stimulus provided by the EPM. The HPA axis response under

the anxiogenic conditions provided by the this test was also disrupted, as indicated by the

higher levels of corticosterone observed in cocaine-treated animals.

Cocaine treatment intensified social investigation and decreased defensive

behaviour during the social conflict provided by the resident-intruder paradigm, indicating

that cocaine affected the ability of male rats to properly interact with conspecifics. The

reduced defensive behaviour observed in cocaine-treated male rats, suggests an impaired

risk-assessment, supporting data obtained in the EPM. The neurochemical data showed

that cocaine decreased the dopaminergic activity in response to the social conflict

provided by the resident-intruder paradigm, with the serotonergic response also being

influenced. The neurochemical alterations may have been relevant to the increased social

investigation and reduced fear observed in cocaine-treated male rats.

283

The neurochemical, hormonal and behavioural effects of cocaine administration

observed in this study are likely contribute to an increased vulnerability to drug abuse and

relapse after withdrawal. Again, the obtained results are also relevant from the social

perspective and can be issued to better structure drug abuse prevention in the

adolescence.

284

4.6. References

Adamec R, Head D, Blundell J, Burton P, Berton O (2006) Lasting anxiogenic effects of feline predator stress in mice: sex differences in vulnerability to stress and predicting severity of anxiogenic response from the stress experience. Physiol Behav 88:12-29.

Albert DJ, Dyson EM, Walsh ML (1987) Intermale social aggression: reinstatement in castrated rats by implants of testosterone propionate in the medial hypothalamus. Physiol Behav 39:555-560.

Albert DJ, Walsh ML, Gorzalka BB, Siemens Y, Louie H (1986) Testosterone removal in rats results in a decrease in social aggression and a loss of social dominance. Physiol Behav 36:401-407.

Albrechet-Souza L, Cristina de Carvalho M, Rodrigues Franci C, Brandao ML (2007) Increases in plasma corticosterone and stretched-attend postures in rats naive and previously exposed to the elevated plus-maze are sensitive to the anxiolytic-like effects of midazolam. Horm Behav 52:267-273.

Amat J, Tamblyn JP, Paul ED, Bland ST, Amat P, Foster AC, Watkins LR, Maier SF (2004) Microinjection of urocortin 2 into the dorsal raphe nucleus activates serotonergic neurons and increases extracellular serotonin in the basolateral amygdala. Neuroscience 129:509-519.

Ambrosio E, Sharpe LG, Pilotte NS (1997) Regional binding to corticotropin releasing factor receptors in brain of rats exposed to chronic cocaine and cocaine withdrawal. Synapse 25:272-276.

Andersen SL (2002) Changes in the second messenger cyclic AMP during development may underlie motoric symptoms in attention deficit/hyperactivity disorder (ADHD). Behav Brain Res 130:197-201.

Andersen SL (2003) Trajectories of brain development: point of vulnerability or window of opportunity? Neurosci Biobehav Rev 27:3-18.

Andersen SL, Gazzara RA (1993) The ontogeny of apomorphine-induced alterations of neostriatal dopamine release: effects on spontaneous release. J Neurochem 61:2247-2255.

Andersen SL, Arvanitogiannis A, Pliakas AM, LeBlanc C, Carlezon WA, Jr. (2002) Altered responsiveness to cocaine in rats exposed to methylphenidate during development. Nat Neurosci 5:13-14.

Antkiewicz-Michaluk L, Filip M, Michaluk J, Romanska I, Patsenka A, Papla I, Przegalinski E, Vetulani J (2006) Conditioned rewarding stimulus associated with cocaine self-administration reverses the depression of catecholamine brain systems following cocaine withdrawal in rats. Int J Neuropsychopharmacol 9:37-50.

Arango V, Underwood MD, Mann JJ (2002) Serotonin brain circuits involved in major depression and suicide. Prog Brain Res 136:443-453.

Ardila A, Rosselli M, Strumwasser S (1991) Neuropsychological deficits in chronic cocaine abusers. Int J Neurosci 57:73-79.

Arroyo M, Baker WA, Everitt BJ (2000) Cocaine self-administration in rats differentially alters mRNA levels of the monoamine transporters and striatal neuropeptides. Brain Res Mol Brain Res 83:107-120.

Bach-Mizrachi H, Underwood MD, Kassir SA, Bakalian MJ, Sibille E, Tamir H, Mann JJ, Arango V (2006) Neuronal tryptophan hydroxylase mRNA expression in the human dorsal and median raphe nuclei: major depression and suicide. Neuropsychopharmacology 31:814-824.

Badiani A, Browman KE, Robinson TE (1995) Influence of novel versus home environments on sensitization to the psychomotor stimulant effects of cocaine and amphetamine. Brain Res 674:291-298.

Balopole DC, Hansult CD, Dorph D (1979) Effect of cocaine on food intake in rats. Psychopharmacology (Berl) 64:121-122.

285

Barfield RJ, Busch DE, Wallen K (1972) Gonadal influence on agonistic behavior in the male domestic rat. Horm Behav 3:247-259.

Barroso-Moguel R, Mendez-Armenta M, Villeda-Hernandez J (1994) Testicular lesions by chronic administration of cocaine in rats. J Appl Toxicol 14:37-41.

Basso AM, Vale W, Koob GF, Spina M, Rivier J (1999) Corticotropin-releasing factor antagonist attenuates the "anxiogenic-like" effect in the defensive burying paradigm but not in the elevated plus-maze following chronic cocaine in rats. Psychopharmacology (Berl) 145:21-30.

Batki SL, Manfredi LB, Jacob P, 3rd, Jones RT (1993) Fluoxetine for cocaine dependence in methadone maintenance: quantitative plasma and urine cocaine/ benzoylecgonine concentrations. J Clin Psychopharmacol 13:243-250.

Baumann MH, Rothman RB (1998) Alterations in serotonergic responsiveness during cocaine withdrawal in rats: similarities to major depression in humans. Biol Psychiatry 44:578-591.

Baumann MH, Gendron TM, Becketts KM, Henningfield JE, Gorelick DA, Rothman RB (1995) Effects of intravenous cocaine on plasma cortisol and prolactin in human cocaine abusers. Biol Psychiatry 38:751-755.

Baxter MG, Murray EA (2002) The amygdala and reward. Nat Rev Neurosci 3:563-573.

Bean NJ, Conner R (1978) Central hormonal replacement and home-cage dominance in castrated rats. Horm Behav 11:100-109.

Beatty WW, Katzung VM, Moreland VJ, Nixon SJ (1995) Neuropsychological performance of recently abstinent alcoholics and cocaine abusers. Drug Alcohol Depend 37:247-253.

Bechara A (2001) Neurobiology of decision-making: risk and reward. Semin Clin Neuropsychiatry 6:205-216.

Bechara A, Damasio H, Damasio AR, Lee GP (1999) Different contributions of the human amygdala and ventromedial prefrontal cortex to decision-making. J Neurosci 19:5473-5481.

Ben-Shahar O, Keeley P, Cook M, Brake W, Joyce M, Nyffeler M, Heston R, Ettenberg A (2007) Changes in levels of D1, D2, or NMDA receptors during withdrawal from brief or extended daily access to IV cocaine. Brain Res 1131:220-228.

Berman ME, Tracy JI, Coccaro EF (1997) The serotonin hypothesis of aggression revisited. Clin Psychol Rev 17:651-665.

Bernabeu R, Bevilaqua L, Ardenghi P, Bromberg E, Schmitz P, Bianchin M, Izquierdo I, Medina JH (1997) Involvement of hippocampal cAMP/cAMP-dependent protein kinase signaling pathways in a late memory consolidation phase of aversively motivated learning in rats. Proc Natl Acad Sci U S A 94:7041-7046.

Berry J, van Gorp WG, Herzberg DS, Hinkin C, Boone K, Steinman L, Wilkins JN (1993) Neuropsychological deficits in abstinent cocaine abusers: preliminary findings after two weeks of abstinence. Drug Alcohol Depend 32:231-237.

Berul CI, Harclerode JE (1989) Effects of cocaine hydrochloride on the male reproductive system. Life Sci 45:91-95.

Beyer CE, Steketee JD (2002) Cocaine sensitization: modulation by dopamine D2 receptors. Cereb Cortex 12:526-535.

Bibancos T, Jardim DL, Aneas I, Chiavegatto S (2007) Social isolation and expression of serotonergic neurotransmission-related genes in several brain areas of male mice. Genes Brain Behav 6:529-539.

Blanchard DC, Canteras NS, Markham CM, Pentkowski NS, Blanchard RJ (2005) Lesions of structures showing FOS expression to cat presentation: effects on responsivity to a Cat, Cat odor, and nonpredator threat. Neurosci Biobehav Rev 29:1243-1253.

286

Bolanos CA, Barrot M, Berton O, Wallace-Black D, Nestler EJ (2003) Methylphenidate treatment during pre- and periadolescence alters behavioral responses to emotional stimuli at adulthood. Biol Psychiatry 54:1317-1329.

Bolla KI, Eldreth DA, London ED, Kiehl KA, Mouratidis M, Contoreggi C, Matochik JA, Kurian V, Cadet JL, Kimes AS, Funderburk FR, Ernst M (2003) Orbitofrontal cortex dysfunction in abstinent cocaine abusers performing a decision-making task. Neuroimage 19:1085-1094.

Brandon CL, Marinelli M, Baker LK, White FJ (2001) Enhanced reactivity and vulnerability to cocaine following methylphenidate treatment in adolescent rats. Neuropsychopharmacology 25:651-661.

Breiter HC, Gollub RL, Weisskoff RM, Kennedy DN, Makris N, Berke JD, Goodman JM, Kantor HL, Gastfriend DR, Riorden JP, Mathew RT, Rosen BR, Hyman SE (1997) Acute effects of cocaine on human brain activity and emotion. Neuron 19:591-611.

Briand LA, Flagel SB, Garcia-Fuster MJ, Watson SJ, Akil H, Sarter M, Robinson TE (2008) Persistent alterations in cognitive function and prefrontal dopamine D2 receptors following extended, but not limited, access to self-administered cocaine. Neuropsychopharmacology 33:2969-2980.

Brown GL, Ebert MH, Goyer PF, Jimerson DC, Klein WJ, Bunney WE, Goodwin FK (1982) Aggression, suicide, and serotonin: relationships to CSF amine metabolites. Am J Psychiatry 139:741-746.

Burchett SA, Bannon MJ (1997) Serotonin, dopamine and norepinephrine transporter mRNAs: heterogeneity of distribution and response to 'binge' cocaine administration. Brain Res Mol Brain Res 49:95-102.

Buydens-Branchey L, Branchey M, Fergeson P, Hudson J, McKernin C (1997) Craving for cocaine in addicted users. Role of serotonergic mechanisms. Am J Addict 6:65-73.

Buydens-Branchey L, Branchey M, Hudson J, Rothman M, Fergeson P, McKernin C (1998) Effect of fenfluramine challenge on cocaine craving in addicted male users. Am J Addict 7:142-155.

Calvo N, Volosin M (2001) Glucocorticoid and mineralocorticoid receptors are involved in the facilitation of anxiety-like response induced by restraint. Neuroendocrinology 73:261-271.

Cao J, Lotfipour S, Loughlin SE, Leslie FM (2007) Adolescent maturation of cocaine-sensitive neural mechanisms. Neuropsychopharmacology 32:2279-2289.

Carlezon WA, Jr., Konradi C (2004) Understanding the neurobiological consequences of early exposure to psychotropic drugs: linking behavior with molecules. Neuropharmacology 47 Suppl 1:47-60.

Carobrez AP, Bertoglio LJ (2005) Ethological and temporal analyses of anxiety-like behavior: the elevated plus-maze model 20 years on. Neurosci Biobehav Rev 29:1193-1205.

Carvalho MC, Albrechet-Souza L, Masson S, Brandao ML (2005) Changes in the biogenic amine content of the prefrontal cortex, amygdala, dorsal hippocampus, and nucleus accumbens of rats submitted to single and repeated sessions of the elevated plus-maze test. Braz J Med Biol Res 38:1857-1866.

Caster JM, Walker QD, Kuhn CM (2005) Enhanced behavioral response to repeated-dose cocaine in adolescent rats. Psychopharmacology (Berl) 183:218-225.

Catlow BJ, Kirstein CL (2005) Heightened cocaine-induced locomotor activity in adolescent compared to adult female rats. J Psychopharmacol 19:443-447.

Catlow BJ, Kirstein CL (2007) Cocaine during adolescence enhances dopamine in response to a natural reinforcer. Neurotoxicol Teratol 29:57-65.

Cerruti C, Pilotte NS, Uhl G, Kuhar MJ (1994) Reduction in dopamine transporter mRNA after cessation of repeated cocaine administration. Brain Res Mol Brain Res 22:132-138.

Chefer VI, Moron JA, Hope B, Rea W, Shippenberg TS (2000) Kappa-opioid receptor activation prevents alterations in mesocortical dopamine neurotransmission that occur during abstinence from cocaine. Neuroscience 101:619-627.

287

Chen CJ, Vandenbergh JG (1994) Effect of chronic cocaine on reproduction in female house mice. Pharmacol Biochem Behav 48:909-913.

Chermack ST, Blow FC (2002) Violence among individuals in substance abuse treatment: the role of alcohol and cocaine consumption. Drug Alcohol Depend 66:29-37.

Cocores JA, Dackis CA, Gold MS (1986) Sexual dysfunction secondary to cocaine abuse in two patients. J Clin Psychiatry 47:384-385.

Coffey SF, Dansky BS, Carrigan MH, Brady KT (2000) Acute and protracted cocaine abstinence in an outpatient population: a prospective study of mood, sleep and withdrawal symptoms. Drug Alcohol Depend 59:277-286.

Collins SL, Izenwasser S (2002) Cocaine differentially alters behavior and neurochemistry in periadolescent versus adult rats. Brain Res Dev Brain Res 138:27-34.

Compan V, Segu L, Buhot MC, Daszuta A (1998) Selective increases in serotonin 5-HT1B/1D and 5-HT2A/2C binding sites in adult rat basal ganglia following lesions of serotonergic neurons. Brain Res 793:103-111.

Cooper SJ, van der Hoek GA (1993) Cocaine: a microstructural analysis of its effects on feeding and associated behaviour in the rat. Brain Res 608:45-51.

Creese I, Iversen SD (1974) The role of forebrain dopamine systems in amphetamine induced stereotyped behavior in the rat. Psychopharmacologia 39:345-357.

Cunningham KA, Paris JM, Goeders NE (1992) Chronic cocaine enhances serotonin autoregulation and serotonin uptake binding. Synapse 11:112-123.

Dackis CA, Gold MS (1985) New concepts in cocaine addiction: the dopamine depletion hypothesis. Neurosci Biobehav Rev 9:469-477.

Dalla C, Antoniou K, Kokras N, Drossopoulou G, Papathanasiou G, Bekris S, Daskas S, Papadopoulou-Daifoti Z (2008) Sex differences in the effects of two stress paradigms on dopaminergic neurotransmission. Physiol Behav 93:595-605.

Dalley JW, Theobald DE, Pereira EA, Li PM, Robbins TW (2002) Specific abnormalities in serotonin release in the prefrontal cortex of isolation-reared rats measured during behavioural performance of a task assessing visuospatial attention and impulsivity. Psychopharmacology (Berl) 164:329-340.

Darmani NA, Hadfield MG, Carter WH, Jr., Martin BR (1990) Acute and chronic effects of cocaine on isolation-induced aggression in mice. Psychopharmacology (Berl) 102:37-40.

Davidson C, Ellinwood EH, Lee TH (2000) Altered sensitivity of dopamine autoreceptors in rat accumbens 1 and 7 days after intermittent or continuous cocaine withdrawal. Brain Res Bull 51:89-93.

De Almeida RM, Lucion AB (1997) 8-OH-DPAT in the median raphe, dorsal periaqueductal gray and corticomedial amygdala nucleus decreases, but in the medial septal area it can increase maternal aggressive behavior in rats. Psychopharmacology (Berl) 134:392-400.

de Boer SF, Koolhaas JM (2005) 5-HT1A and 5-HT1B receptor agonists and aggression: a pharmacological challenge of the serotonin deficiency hypothesis. Eur J Pharmacol 526:125-139.

de Boer SF, Lesourd M, Mocaer E, Koolhaas JM (1999) Selective antiaggressive effects of alnespirone in resident-intruder test are mediated via 5-hydroxytryptamine1A receptors: A comparative pharmacological study with 8-hydroxy-2-dipropylaminotetralin, ipsapirone, buspirone, eltoprazine, and WAY-100635. J Pharmacol Exp Ther 288:1125-1133.

de Kloet ER (2003) Hormones, brain and stress. Endocr Regul 37:51-68.

DeLeon KR, Grimes JM, Connor DF, Melloni RH, Jr. (2002) Adolescent cocaine exposure and offensive aggression: involvement of serotonin neural signaling and innervation in male Syrian hamsters. Behav Brain Res 133:211-220.

Delville Y, Mansour KM, Ferris CF (1996) Testosterone facilitates aggression by modulating vasopressin receptors in the hypothalamus. Physiol Behav 60:25-29.

288

Denison ME, Paredes A, Booth JB (1997) Alcohol and cocaine interactions and aggressive behaviors. Recent Dev Alcohol 13:283-303.

Deroche V, Marinelli M, Le Moal M, Piazza PV (1997) Glucocorticoids and behavioral effects of psychostimulants. II: cocaine intravenous self-administration and reinstatement depend on glucocorticoid levels. J Pharmacol Exp Ther 281:1401-1407.

Elman I, Lukas SE, Karlsgodt KH, Gasic GP, Breiter HC (2003) Acute cortisol administration triggers craving in individuals with cocaine dependence. Psychopharmacol Bull 37:84-89.

Emley GS, Hutchinson RR (1983) Unique influences of ten drugs upon post-shock biting attack and pre-shock manual responding. Pharmacol Biochem Behav 19:5-12.

Erb S, Shaham Y, Stewart J (1998) The role of corticotropin-releasing factor and corticosterone in stress- and cocaine-induced relapse to cocaine seeking in rats. J Neurosci 18:5529-5536.

Erb S, Kayyali H, Romero K (2006) A study of the lasting effects of cocaine pre-exposure on anxiety-like behaviors under baseline conditions and in response to central injections of corticotropin-releasing factor. Pharmacol Biochem Behav 85:206-213.

Estelles, Aguilar MA, Miñarro J, Lluch J, Rodríguez-Arias M (2007) Cocaine exposure during adolescence affects anxiety in adult mice. Brain Res Bull 71:393-403.

Estelles J, Rodriguez-Arias M, Maldonado C, Aguilar MA, Minarro J (2005) Prenatal cocaine exposure alters spontaneous and cocaine-induced motor and social behaviors. Neurotoxicol Teratol 27:449-457.

Everitt BJ, Wolf ME (2002) Psychomotor stimulant addiction: a neural systems perspective. J Neurosci 22:3312-3320.

Fals-Stewart W, Golden J, Schumacher JA (2003) Intimate partner violence and substance use: a longitudinal day-to-day examination. Addict Behav 28:1555-1574.

Fanselow MS, LeDoux JE (1999) Why we think plasticity underlying Pavlovian fear conditioning occurs in the basolateral amygdala. Neuron 23:229-232.

Feltenstein MW, See RE (2007) Plasma progesterone levels and cocaine-seeking in freely cycling female rats across the estrous cycle. Drug Alcohol Depend 89:183-189.

Ferre S, Cortes R, Artigas F (1994) Dopaminergic regulation of the serotonergic raphe-striatal pathway: microdialysis studies in freely moving rats. J Neurosci 14:4839-4846.

Ferris CF, Stolberg T, Delville Y (1999) Serotonin regulation of aggressive behavior in male golden hamsters (Mesocricetus auratus). Behav Neurosci 113:804-815.

File SE, Seth P (2003) A review of 25 years of the social interaction test. Eur J Pharmacol 463:35-53.

Filibeck U, Cabib S, Castellano C, Puglisi-Allegra S (1988) Chronic cocaine enhances defensive behaviour in the laboratory mouse: involvement of D2 dopamine receptors. Psychopharmacology (Berl) 96:437-441.

Floresco SB, Tse MT (2007) Dopaminergic regulation of inhibitory and excitatory transmission in the basolateral amygdala-prefrontal cortical pathway. J Neurosci 27:2045-2057.

Fontana DJ, Commissaris RL (1989) Effects of cocaine on conflict behavior in the rat. Life Sci 45:819-827.

Frey U, Schroeder H, Matthies H (1990) Dopaminergic antagonists prevent long-term maintenance of posttetanic LTP in the CA1 region of rat hippocampal slices. Brain Res 522:69-75.

Gammie SC, Nelson RJ (2001) cFOS and pCREB activation and maternal aggression in mice. Brain Res 898:232-241.

Gao WY, Lee TH, King GR, Ellinwood EH (1998) Alterations in baseline activity and quinpirole sensitivity in putative dopamine neurons in the substantia nigra and ventral tegmental area after withdrawal from cocaine pretreatment. Neuropsychopharmacology 18:222-232.

289

Gargiulo PA, Viana MB, Graeff FG, Silva MA, Tomaz C (1996) Effects of anxiety and memory of systemic and intra-amygdala injection of 5-HT3 receptor antagonist BRL 46470A. Neuropsychobiology 33:189-195.

Garlow SJ, Purselle D, D'Orio B (2003) Cocaine use disorders and suicidal ideation. Drug Alcohol Depend 70:101-104.

Gasbarri A, Sulli A, Innocenzi R, Pacitti C, Brioni JD (1996) Spatial memory impairment induced by lesion of the mesohippocampal dopaminergic system in the rat. Neuroscience 74:1037-1044.

Gawin FH (1991) Cocaine addiction: psychology and neurophysiology. Science 251:1580-1586.

Gawin FH, Kleber HD (1986) Abstinence symptomatology and psychiatric diagnosis in cocaine abusers. Clinical observations. Arch Gen Psychiatry 43:107-113.

Gawin FH, Kleber HD, Byck R, Rounsaville BJ, Kosten TR, Jatlow PI, Morgan C (1989) Desipramine facilitation of initial cocaine abstinence. Arch Gen Psychiatry 46:117-121.

George VK, Li H, Teloken C, Grignon DJ, Lawrence WD, Dhabuwala CB (1996) Effects of long-term cocaine exposure on spermatogenesis and fertility in peripubertal male rats. J Urol 155:327-331.

Gerra G, Zaimovic A, Avanzini P, Chittolini B, Giucastro G, Caccavari R, Palladino M, Maestri D, Monica C, Delsignore R, Brambilla F (1997) Neurotransmitter-neuroendocrine responses to experimentally induced aggression in humans: influence of personality variable. Psychiatry Res 66:33-43.

Giorgetti M, Zhdanova IV (2000) Chronic cocaine treatment induces dysregulation in the circadian pattern of rats' feeding behavior. Brain Res 877:170-175.

Gomez F, Dallman MF (2001) Manipulation of androgens causes different energetic responses to cold in 60- and 40-day-old male rats. Am J Physiol Regul Integr Comp Physiol 280:R262-273.

Gomez F, Houshyar H, Dallman MF (2002) Marked regulatory shifts in gonadal, adrenal, and metabolic system responses to repeated restraint stress occur within a 3-week period in pubertal male rats. Endocrinology 143:2852-2862.

Gordon MK, Rosen JB (1999) Lasting effect of repeated cocaine administration on acoustic and fear-potentiated startle in rats. Psychopharmacology (Berl) 144:1-7.

Gregg TR, Siegel A (2001) Brain structures and neurotransmitters regulating aggression in cats: implications for human aggression. Prog Neuropsychopharmacol Biol Psychiatry 25:91-140.

Gupta GL, Rana AC (2007) Protective effect of Withania somnifera dunal root extract against protracted social isolation induced behavior in rats. Indian J Physiol Pharmacol 51:345-353.

Hadfield MG, Nugent EA, Mott DE (1982) Cocaine increases isolation-induced fighting in mice. Pharmacol Biochem Behav 16:359-360.

Haj-Dahmane S (2001) D2-like dopamine receptor activation excites rat dorsal raphe 5-HT neurons in vitro. Eur J Neurosci 14:125-134.

Halasz J, Liposits Z, Kruk MR, Haller J (2002) Neural background of glucocorticoid dysfunction-induced abnormal aggression in rats: involvement of fear- and stress-related structures. Eur J Neurosci 15:561-569.

Hall FS (1998) Social deprivation of neonatal, adolescent, and adult rats has distinct neurochemical and behavioral consequences. Crit Rev Neurobiol 12:129-162.

Haller J, Kruk MR (2006) Normal and abnormal aggression: human disorders and novel laboratory models. Neurosci Biobehav Rev 30:292-303.

Haller J, van de Schraaf J, Kruk MR (2001) Deviant forms of aggression in glucocorticoid hyporeactive rats: a model for 'pathological' aggression? J Neuroendocrinol 13:102-107.

290

Haller J, Halasz J, Mikics E, Kruk MR (2004) Chronic glucocorticoid deficiency-induced abnormal aggression, autonomic hypoarousal, and social deficit in rats. J Neuroendocrinol 16:550-557.

Hammack SE, Schmid MJ, LoPresti ML, Der-Avakian A, Pellymounter MA, Foster AC, Watkins LR, Maier SF (2003) Corticotropin releasing hormone type 2 receptors in the dorsal raphe nucleus mediate the behavioral consequences of uncontrollable stress. J Neurosci 23:1019-1025.

Handelsman L, Rosenblum A, Palij M, Magura S, Foote J, Lovejoy M, Stimmel B (1997) Bromocriptine for cocaine dependence. A controlled clinical trial. Am J Addict 6:54-64.

Harrison RJ, Connor DF, Nowak C, Melloni RH, Jr. (2000) Chronic low-dose cocaine treatment during adolescence facilitates aggression in hamsters. Physiol Behav 69:555-562.

Hayes RJ, Gardner EL (2004) The basolateral complex of the amygdala mediates the modulation of intracranial self-stimulation threshold by drug-associated cues. Eur J Neurosci 20:273-280.

Higgins GA, Jones BJ, Oakley NR (1992) Effect of 5-HT1A receptor agonists in two models of anxiety after dorsal raphe injection. Psychopharmacology (Berl) 106:261-267.

Higgins GA, Bradbury AJ, Jones BJ, Oakley NR (1988) Behavioural and biochemical consequences following activation of 5HT1-like and GABA receptors in the dorsal raphe nucleus of the rat. Neuropharmacology 27:993-1001.

Higgins GA, Jones BJ, Oakley NR, Tyers MB (1991) Evidence that the amygdala is involved in the disinhibitory effects of 5-HT3 receptor antagonists. Psychopharmacology (Berl) 104:545-551.

Higley JD, Mehlman PT, Taub DM, Higley SB, Suomi SJ, Vickers JH, Linnoila M (1992) Cerebrospinal fluid monoamine and adrenal correlates of aggression in free-ranging rhesus monkeys. Arch Gen Psychiatry 49:436-441.

Hoff AL, Riordan H, Morris L, Cestaro V, Wieneke M, Alpert R, Wang GJ, Volkow N (1996) Effects of crack cocaine on neurocognitive function. Psychiatry Res 60:167-176.

Hoffman BJ, Hansson SR, Mezey E, Palkovits M (1998) Localization and dynamic regulation of biogenic amine transporters in the mammalian central nervous system. Front Neuroendocrinol 19:187-231.

Hogg S (1996) A review of the validity and variability of the elevated plus-maze as an animal model of anxiety. Pharmacol Biochem Behav 54:21-30.

Huang YY, Kandel ER (1995) D1/D5 receptor agonists induce a protein synthesis-dependent late potentiation in the CA1 region of the hippocampus. Proc Natl Acad Sci U S A 92:2446-2450.

Jacobsen LK, Giedd JN, Kreek MJ, Gottschalk C, Kosten TR (2001) Quantitative medial temporal lobe brain morphology and hypothalamic-pituitary-adrenal axis function in cocaine dependence: a preliminary report. Drug Alcohol Depend 62:49-56.

Jacobsen LK, Staley JK, Malison RT, Zoghbi SS, Seibyl JP, Kosten TR, Innis RB (2000) Elevated central serotonin transporter binding availability in acutely abstinent cocaine-dependent patients. American Journal of Psychiatry 157:1134-1140.

Johansson AK, Hansen S (2002) Novelty seeking and harm avoidance in relation to alcohol drinking in intact rats and following axon-sparing lesions to the amygdala and ventral striatum. Alcohol Alcohol 37:147-156.

Johns JM, Means MJ, Bass EW, Means LW, Zimmerman LI, McMillen BA (1994) Prenatal exposure to cocaine: effects on aggression in Sprague-Dawley rats. Dev Psychobiol 27:227-239.

Jones SR, Lee TH, Wightman RM, Ellinwood EH (1996) Effects of intermittent and continuous cocaine administration on dopamine release and uptake regulation in the striatum: in vitro voltammetric assessment. Psychopharmacology (Berl) 126:331-338.

291

Kalivas PW, McFarland K (2003) Brain circuitry and the reinstatement of cocaine-seeking behavior. Psychopharmacology (Berl) 168:44-56.

Kampman K, Volpicelli JR, Alterman A, Cornish J, Weinrieb R, Epperson L, Sparkman T, O'Brien CP (1996) Amantadine in the early treatment of cocaine dependence: a double-blind, placebo-controlled trial. Drug Alcohol Depend 41:25-33.

Kampman KM, Rukstalis M, Pettinati H, Muller E, Acosta T, Gariti P, Ehrman R, O'Brien CP (2000) The combination of phentermine and fenfluramine reduced cocaine withdrawal symptoms in an open trial. J Subst Abuse Treat 19:77-79.

Karoum F, Egan MF, Wyatt RJ (1994) Selective reduction in dopamine turnover in the rat frontal cortex and hypothalamus during withdrawal from repeated cocaine exposure. Eur J Pharmacol 254:127-132.

Kavoussi R, Armstead P, Coccaro E (1997) The neurobiology of impulsive aggression. Psychiatr Clin North Am 20:395-403.

Kelley AE (2004) Memory and addiction: shared neural circuitry and molecular mechanisms. Neuron 44:161-179.

Kimmel HL, Carroll FI, Kuhar MJ (2003) Withdrawal from repeated cocaine alters dopamine transporter protein turnover in the rat striatum. J Pharmacol Exp Ther 304:15-21.

Kimura T, Ishihara K, Ozawa K, Sasa M (2004) Augmentation of serotonin-induced inhibition of neuronal activity in the hippocampus following repeated treatment with methamphetamine. Ann N Y Acad Sci 1025:242-247.

King GR, Kuhn C, Ellinwood EH, Jr. (1993a) Dopamine efflux during withdrawal from continuous or intermittent cocaine. Psychopharmacology (Berl) 111:179-184.

King TS, Canez MS, Gaskill S, Javors MA, Schenken RS (1993b) Chronic cocaine disruption of estrous cyclicity in the rat: dose-dependent effects. J Pharmacol Exp Ther 264:29-34.

Kitahama K, Nagatsu I, Geffard M, Maeda T (2000) Distribution of dopamine-immunoreactive fibers in the rat brainstem. J Chem Neuroanat 18:1-9.

Kogan JH, Frankland PW, Silva AJ (2000) Long-term memory underlying hippocampus-dependent social recognition in mice. Hippocampus 10:47-56.

Koob G, Sanna P, Bloom F (1998) Neuroscience of addiction. Neuron 21:467-476.

Koob GF, Le Moal M (1997) Drug abuse: hedonic homeostatic dysregulation. Science 278:52-58.

Koolhaas JM, Schuurman T, Wiepkema PR (1980) The organization of intraspecific agonistic behaviour in the rat. Prog Neurobiol 15:247-268.

Korte SM, de Boer SF, de Kloet ER, Bohus B (1995) Anxiolytic-like effects of selective mineralocorticoid and glucocorticoid antagonists on fear-enhanced behavior in the elevated plus-maze. Psychoneuroendocrinology 20:385-394.

Kosten TA, Zhang XY, Kehoe P (2003) Chronic neonatal isolation stress enhances cocaine-induced increases in ventral striatal dopamine levels in rat pups. Brain Res Dev Brain Res 141:109-116.

Krimer LS, Jakab RL, Goldman-Rakic PS (1997) Quantitative three-dimensional analysis of the catecholaminergic innervation of identified neurons in the macaque prefrontal cortex. J Neurosci 17:7450-7461.

Kruesi MJ, Rapoport JL, Hamburger S, Hibbs E, Potter WZ, Lenane M, Brown GL (1990) Cerebrospinal fluid monoamine metabolites, aggression, and impulsivity in disruptive behavior disorders of children and adolescents. Arch Gen Psychiatry 47:419-426.

Kruk MR, Halasz J, Meelis W, Haller J (2004) Fast positive feedback between the adrenocortical stress response and a brain mechanism involved in aggressive behavior. Behav Neurosci 118:1062-1070.

Kyes RC, Botchin MB, Kaplan JR, Manuck SB, Mann JJ (1995) Aggression and brain serotonergic responsivity: response to slides in male macaques. Physiol Behav 57:205-208.

292

Laurier LG, Corrigall WA, George SR (1994) Dopamine receptor density, sensitivity and mRNA levels are altered following self-administration of cocaine in the rat. Brain Res 634:31-40.

Laviola G, Wood RD, Kuhn C, Francis R, Spear LP (1995) Cocaine sensitization in periadolescent and adult rats. J Pharmacol Exp Ther 275:345-357.

LeDoux JE (2000) Emotion circuits in the brain. Annu Rev Neurosci 23:155-184.

Letchworth SR, Daunais JB, Hedgecock AA, Porrino LJ (1997) Effects of chronic cocaine administration on dopamine transporter mRNA and protein in the rat. Brain Res 750:214-222.

Letchworth SR, Sexton T, Childers SR, Vrana KE, Vaughan RA, Davies HM, Porrino LJ (1999) Regulation of rat dopamine transporter mRNA and protein by chronic cocaine administration. J Neurochem 73:1982-1989.

Levy AD, Li Q, Alvarez Sanz MC, Rittenhouse PA, Kerr JE, Van de Kar LD (1992a) Neuroendocrine responses to cocaine do not exhibit sensitization following repeated cocaine exposure. Life Sci 51:887-897.

Levy AD, Li Q, Alvarez Sanz MC, Rittenhouse PA, Brownfield MS, Van de Kar LD (1992b) Repeated cocaine modifies the neuroendocrine responses to the 5-HT1C/5-HT2 receptor agonist DOI. Eur J Pharmacol 221:121-127.

Lewis DA (1997) Development of the prefrontal cortex during adolescence: insights into vulnerable neural circuits in schizophrenia. Neuropsychopharmacology 16:385-398.

Lin Y, Ter Horst GJ, Wichmann R, Bakker P, Liu A, Li X, Westenbroek C (2009) Sex differences in the effects of acute and chronic stress and recovery after long-term stress on stress-related brain regions of rats. Cereb Cortex 19:1978-1989.

Linnoila M, Virkkunen M, Scheinin M, Nuutila A, Rimon R, Goodwin FK (1983) Low cerebrospinal fluid 5-hydroxyindoleacetic acid concentration differentiates impulsive from nonimpulsive violent behavior. Life Sci 33:2609-2614.

Linnoila VM, Virkkunen M (1992) Aggression, suicidality, and serotonin. J Clin Psychiatry 53 Suppl:46-51.

Lira A, Zhou M, Castanon N, Ansorge MS, Gordon JA, Francis JH, Bradley-Moore M, Lira J, Underwood MD, Arango V, Kung HF, Hofer MA, Hen R, Gingrich JA (2003) Altered depression-related behaviors and functional changes in the dorsal raphe nucleus of serotonin transporter-deficient mice. Biol Psychiatry 54:960-971.

Lista A, Blier P, de Montigny C (1990) Benzodiazepine receptors modulate 5-hydroxytryptamine neurotransmission in the rat hippocampus: in vivo electrophysiological evidence. J Pharmacol Exp Ther 254:318-323.

Long SF, Wilson MC, Sufka KJ, Davis WM (1996) The effects of cocaine and nandrolone co-administration on aggression in male rats. Prog Neuropsychopharmacol Biol Psychiatry 20:839-856.

Lucas LR, Schlussman SD, Ho A, McEwen BS, Kreek MJ (1997) Novelty-induced locomoter activity in Long-Evans rats pre- and post-chronic 'binge'-pattern cocaine treatment. Neurosci Lett 237:25-28.

Luiten PG, Koolhaas JM, de Boer S, Koopmans SJ (1985) The cortico-medial amygdala in the central nervous system organization of agonistic behavior. Brain Res 332:283-297.

Macri S, Adriani W, Chiarotti F, Laviola G (2002) Risk taking during exploration of a plus-maze is greater in adolescent than in juvenile or adult mice. Animal Behaviour 64:541-546.

Maggos CE, Spangler R, Zhou Y, Schlussman SD, Ho A, Kreek MJ (1997) Quantitation of dopamine transporter mRNA in the rat brain: mapping, effects of "binge" cocaine administration and withdrawal. Synapse 26:55-61.

Maier SF, Busch CR, Maswood S, Grahn RE, Watkins LR (1995) The dorsal raphe nucleus is a site of action mediating the behavioral effects of the benzodiazepine receptor inverse agonist DMCM. Behav Neurosci 109:759-766.

293

Maisonneuve IM, Ho A, Kreek MJ (1995) Chronic administration of a cocaine "binge" alters basal extracellular levels in male rats: an in vivo microdialysis study. J Pharmacol Exp Ther 272:652-657.

Majchrzak M, Malendowicz LK (1983) Sex differences in adrenocortical structure and function. XII. Stereologic studies of rat adrenal cortex in the course of maturation. Cell Tissue Res 232:457-469.

Malanga CJ, Riday TT, Carlezon WA, Jr., Kosofsky BE (2008) Prenatal exposure to cocaine increases the rewarding potency of cocaine and selective dopaminergic agonists in adult mice. Biol Psychiatry 63:214-221.

Maldonado AM, Kirstein CL (2005a) Handling alters cocaine-induced activity in adolescent but not adult male rats. Physiol Behav 84:321-326.

Maldonado AM, Kirstein CL (2005b) Cocaine-induced locomotor activity is increased by prior handling in adolescent but not adult female rats. Physiol Behav 86:568-572.

Mann JJ (2003) Neurobiology of suicidal behaviour. Nat Rev Neurosci 4:819-828.

Manrique C, Francois-Bellan AM, Segu L, Becquet D, Hery M, Faudon M, Hery F (1994) Impairment of serotoninergic transmission is followed by adaptive changes in 5HT1B binding sites in the rat suprachiasmatic nucleus. Brain Res 663:93-100.

Manschreck TC, Schneyer ML, Weisstein CC, Laughery J, Rosenthal J, Celada T, Berner J (1990) Freebase cocaine and memory. Compr Psychiatry 31:369-375.

Mansour A, Meador-Woodruff JH, Bunzow JR, Civelli O, Akil H, Watson SJ (1990) Localization of dopamine D2 receptor mRNA and D1 and D2 receptor binding in the rat brain and pituitary: an in situ hybridization-receptor autoradiographic analysis. J Neurosci 10:2587-2600.

Mantsch JR, Goeders NE (1999) Ketoconazole blocks the stress-induced reinstatement of cocaine-seeking behavior in rats: relationship to the discriminative stimulus effects of cocaine. Psychopharmacology (Berl) 142:399-407.

Mantsch JR, Schlussman SD, Ho A, Kreek MJ (2000) Effects of cocaine self-administration on plasma corticosterone and prolactin in rats. J Pharmacol Exp Ther 294:239-247.

Maren S (2001) Neurobiology of Pavlovian fear conditioning. Annu Rev Neurosci 24:897-931.

Marinelli M, Piazza PV (2002) Interaction between glucocorticoid hormones, stress and psychostimulant drugs. Eur J Neurosci 16:387-394.

Mash DC, Pablo J, Ouyang Q, Hearn WL, Izenwasser S (2002) Dopamine transport function is elevated in cocaine users. J Neurochem 81:292-300.

Matsumoto K, Pinna G, Puia G, Guidotti A, Costa E (2005) Social isolation stress-induced aggression in mice: a model to study the pharmacology of neurosteroidogenesis. Stress 8:85-93.

Matsumoto M, Yoshioka M, Togashi H, Ikeda T, Saito H (1996) Functional regulation by dopamine receptors of serotonin release from the rat hippocampus: in vivo microdialysis study. Naunyn Schmiedebergs Arch Pharmacol 353:621-629.

Matuszewich L, Karney JJ, Carter SR, Janasik SP, O'Brien JL, Friedman RD (2007) The delayed effects of chronic unpredictable stress on anxiety measures. Physiol Behav 90:674-681.

McBurnett K, Lahey BB (1994) Psychophysiological and neuroendocrine correlates of conduct disorder and antisocial behavior in children and adolescents. Prog Exp Pers Psychopathol Res:199-231.

McBurnett K, Lahey BB, Rathouz PJ, Loeber R (2000) Low salivary cortisol and persistent aggression in boys referred for disruptive behavior. Arch Gen Psychiatry 57:38-43.

Mello NK, Mendelson JH (1997) Cocaine's effects on neuroendocrine systems: clinical and preclinical studies. Pharmacol Biochem Behav 57:571-599.

Mello NK, Mendelson JH, Kelly M, Diaz-Migoyo N, Sholar JW (1997) The Effects of Chronic Cocaine Self-Administration on the Menstrual Cycle in Rhesus Monkeys. J Pharmacol Exp Ther 281:70-83.

294

Merline AC, O'Malley PM, Schulenberg JE, Bachman JG, Johnston LD (2004) Substance use among adults 35 years of age: Prevalence, adulthood predictors, and impact of adolescent substance use. American Journal of Public Health 94:96-102.

Miczek KA (1979) A new test for aggression in rats without aversive stimulation: differential effects of d-amphetamine and cocaine. Psychopharmacology (Berl) 60:253-259.

Miczek KA, Hussain S, Faccidomo S (1998) Alcohol-heightened aggression in mice: attenuation by 5-HT1A receptor agonists. Psychopharmacology (Berl) 139:160-168.

Miczek KA, Fish EW, De Bold JF, De Almeida RM (2002) Social and neural determinants of aggressive behavior: pharmacotherapeutic targets at serotonin, dopamine and gamma-aminobutyric acid systems. Psychopharmacology (Berl) 163:434-458.

Mikics E, Kruk MR, Haller J (2004) Genomic and non-genomic effects of glucocorticoids on aggressive behavior in male rats. Psychoneuroendocrinology 29:618-635.

Mikkelsen JD, Soderman A, Kiss A, Mirza N (2005) Effects of benzodiazepines receptor agonists on the hypothalamic-pituitary-adrenocortical axis. Eur J Pharmacol 519:223-230.

Miserendino MJ, Sananes CB, Melia KR, Davis M (1990) Blocking of acquisition but not expression of conditioned fear-potentiated startle by NMDA antagonists in the amygdala. Nature 345:716-718.

Moeller FG, Steinberg JL, Petty F, Fulton M, Cherek DR, Kramer G, Garver DL (1994) Serotonin and impulsive/aggressive behavior in cocaine dependent subjects. Prog Neuropsychopharmacol Biol Psychiatry 18:1027-1035.

Moore TM, Stuart GL, Meehan JC, Rhatigan DL, Hellmuth JC, Keen SM (2008) Drug abuse and aggression between intimate partners: a meta-analytic review. Clin Psychol Rev 28:247-274.

Muller M, Fendt M (2006) Temporary inactivation of the medial and basolateral amygdala differentially affects TMT-induced fear behavior in rats. Behav Brain Res 167:57-62.

Munoz-Abellan C, Andero R, Nadal R, Armario A (2008) Marked dissociation between hypothalamic-pituitary-adrenal activation and long-term behavioral effects in rats exposed to immobilization or cat odor. Psychoneuroendocrinology 33:1139-1150.

Mutschler NH, Miczek KA (1998) Withdrawal from i.v. cocaine "binges" in rats: ultrasonic distress calls and startle. Psychopharmacology (Berl) 135:161-168.

Myers DA, Gibson M, Schulkin J, Greenwood Van-Meerveld B (2005) Corticosterone implants to the amygdala and type 1 CRH receptor regulation: effects on behavior and colonic sensitivity. Behav Brain Res 161:39-44.

Nader K, Schafe GE, Le Doux JE (2000) Fear memories require protein synthesis in the amygdala for reconsolidation after retrieval. Nature 406:722-726.

Nestler EJ (2001) Molecular basis of long-term plasticity underlying addiction. Nat Rev Neurosci 2:119-128.

Nestler EJ, Barrot M, DiLeone RJ, Eisch AJ, Gold SJ, Monteggia LM (2002) Neurobiology of depression. Neuron 34:13-25.

Niesink RJ, van Ree JM (1982) Short-term isolation increases social interactions of male rats: a parametric analysis. Physiol Behav 29:819-825.

Nogueira L, Kalivas PW, Lavin A (2006) Long-term neuroadaptations produced by withdrawal from repeated cocaine treatment: role of dopaminergic receptors in modulating cortical excitability. J Neurosci 26:12308-12313.

Olivier B (2004) Serotonin and aggression. Ann N Y Acad Sci 1036:382-392.

Otmakhova NA, Lisman JE (1996) D1/D5 dopamine receptor activation increases the magnitude of early long-term potentiation at CA1 hippocampal synapses. J Neurosci 16:7478-7486.

Packard MG, White NM (1991) Dissociation of hippocampus and caudate nucleus memory systems by posttraining intracerebral injection of dopamine agonists. Behav Neurosci 105:295-306.

295

Parsons LH, Smith AD, Justice JB, Jr. (1991) Basal extracellular dopamine is decreased in the rat nucleus accumbens during abstinence from chronic cocaine. Synapse 9:60-65.

Parsons LH, Koob GF, Weiss F (1995) Serotonin dysfunction in the nucleus accumbens of rats during withdrawal after unlimited access to intravenous cocaine. J Pharmacol Exp Ther 274:1182-1191.

Pellow S, Chopin P, File SE, Briley M (1985) Validation of open:closed arm entries in an elevated plus-maze as a measure of anxiety in the rat. J Neurosci Methods 14:149-167.

Persico AM, Schindler CW, Zaczek R, Brannock MT, Uhl GR (1995) Brain transcription factor gene expression, neurotransmitter levels, and novelty response behaviors: alterations during rat amphetamine withdrawal and following chronic injection stress. Synapse 19:212-227.

Peyron C, Luppi PH, Kitahama K, Fort P, Hermann DM, Jouvet M (1995) Origin of the dopaminergic innervation of the rat dorsal raphe nucleus. Neuroreport 6:2527-2531.

Philpot RM, Kirstein CL (1999) Repeated cocaine exposure: effects on catecholamines in the nucleus accumbens septi of periadolescent animals. Pharmacol Biochem Behav 62:465-472.

Pierce RC, Kalivas PW (1997) A circuitry model of the expression of behavioral sensitization to amphetamine-like psychostimulants. Brain Res Brain Res Rev 25:192-216.

Pollack MH, Rosenbaum JF (1991) Fluoxetine treatment of cocaine abuse in heroin addicts. J Clin Psychiatry 52:31-33.

Post RM, Lockfeld A, Squillace KM, Contel NR (1981) Drug-environment interaction: context dependency of cocaine-induced behavioral sensitization. Life Sci 28:755-760.

Przegalinski E, Czepiel K, Nowak E, Dlaboga D, Filip M (2003) Withdrawal from chronic cocaine up-regulates 5-HT1B receptors in the rat brain. Neurosci Lett 351:169-172.

Rademacher DJ, Schuyler AL, Kruschel CK, Steinpreis RE (2002) Effects of cocaine and putative atypical antipsychotics on rat social behavior: an ethopharmacological study. Pharmacol Biochem Behav 73:769-778.

Ricci LA, Grimes JM, Melloni RH, Jr. (2004) Serotonin type 3 receptors modulate the aggression-stimulating effects of adolescent cocaine exposure in Syrian hamsters (Mesocricetus auratus). Behav Neurosci 118:1097-1110.

Rilke O, Freier D, Jahkel M, Oehler J (1998) Dynamic alterations of serotonergic metabolism and receptors during social isolation of low- and high-active mice. Pharmacol Biochem Behav 59:891-896.

Rios-Bedoya CF, Wilcox HC, Piazza M, Anthony JC (2008) Children taking risks: the association with cocaine and other drug use by young adulthood. Addict Behav 33:1154-1161.

Rivier C, Lee S (1994) Stimulatory effect of cocaine on ACTH secretion: role of the hypothalamus. Mol Cell Neurosci 5:189-195.

Robbins TW, Everitt BJ (1996) Neurobehavioural mechanisms of reward and motivation. Curr Opin Neurobiol 6:228-236.

Robbins TW, Cador M, Taylor JR, Everitt BJ (1989) Limbic-striatal interactions in reward-related processes. Neurosci Biobehav Rev 13:155-162.

Roberts DC, Koob GF, Klonoff P, Fibiger HC (1980) Extinction and recovery of cocaine self-administration following 6-hydroxydopamine lesions of the nucleus accumbens. Pharmacol Biochem Behav 12:781-787.

Rodgers RJ, Cole JC (1993) Influence of social isolation, gender, strain, and prior novelty on plus-maze behaviour in mice. Physiol Behav 54:729-736.

Rodgers RJ, Johnson NJ (1995) Factor analysis of spatiotemporal and ethological measures in the murine elevated plus-maze test of anxiety. Pharmacol Biochem Behav 52:297-303.

Rodgers RJ, Haller J, Holmes A, Halasz J, Walton TJ, Brain PF (1999) Corticosterone response to the plus-maze: high correlation with risk assessment in rats and mice. Physiol Behav 68:47-53.

296

Rose RM, Holaday JW, Bernstein IS (1971) Plasma testosterone, dominance rank and aggressive behaviour in male rhesus monkeys. Nature 231:366-368.

Santucci AC, Capodilupo S, Bernstein J, Gomez-Ramirez M, Milefsky R, Mitchell H (2004) Cocaine in adolescent rats produces residual memory impairments that are reversible with time. Neurotoxicol Teratol 26:651-661.

Sarnyai Z, Dhabhar FS, McEwen BS, Kreek MJ (1998) Neuroendocrine-related effects of long-term, 'binge' cocaine administration: Diminished individual differences in stress-induced corticosterone response. Neuroendocrinology 68:334-344.

Sarnyai Z, Vecsernyés M, Julesz J, Bíró E, Gardi J, Telegdy G (1995) Brain corticotropin-releasing factor mediates 'anxiety-like' behavior induced by cocaine withdrawal in rats. Brain Res 675:89-97.

Satel SL, Price LH, Palumbo JM, McDougle CJ, Krystal JH, Gawin F, Charney DS, Heninger GR, Kleber HD (1991) Clinical phenomenology and neurobiology of cocaine abstinence: a prospective inpatient study. Am J Psychiatry 148:1712-1716.

Schreiber R, De Vry J (1993) Neuronal circuits involved in the anxiolytic effects of the 5-HT1A receptor agonists 8-OH-DPAT ipsapirone and buspirone in the rat. Eur J Pharmacol 249:341-351.

Schuurman T (1980) Hormonal correlates of agonistic behavior in adult male rats. Prog Brain Res 53:415-420.

Seamans JK, Yang CR (2004) The principal features and mechanisms of dopamine modulation in the prefrontal cortex. Prog Neurobiol 74:1-58.

Segal DS, Kuczenski R (1992) Repeated cocaine administration induces behavioral sensitization and corresponding decreased extracellular dopamine responses in caudate and accumbens. Brain Res 577:351-355.

Shaham Y, Erb S, Stewart J (2000) Stress-induced relapse to heroin and cocaine seeking in rats: a review. Brain Research Reviews 33:13-33.

Shalev U, Grimm JW, Shaham Y (2002) Neurobiology of relapse to heroin and cocaine seeking: A review. Pharmacol Rev 54:1-42.

Shepard JD, Barron KW, Myers DA (2000) Corticosterone delivery to the amygdala increases corticotropin-releasing factor mRNA in the central amygdaloid nucleus and anxiety-like behavior. Brain Res 861:288-295.

Shepard JD, Barron KW, Myers DA (2003) Stereotaxic localization of corticosterone to the amygdala enhances hypothalamo-pituitary-adrenal responses to behavioral stress. Brain Res 963:203-213.

Shoal GD, Giancola PR, Kirillova GP (2003) Salivary cortisol, personality, and aggressive behavior in adolescent boys: a 5-year longitudinal study. J Am Acad Child Adolesc Psychiatry 42:1101-1107.

Siciliano D, Smith R (2001) Periadolescent alcohol alters adult behavioral characteristics in the rat. Physiol Behav 74:637-643.

Siegel RK (1982) Cocaine and sexual dysfunction: the curse of mama coca. J Psychoactive Drugs 14:71-74.

Simon NG, Cologer-Clifford A, Lu SF, McKenna SE, Hu S (1998) Testosterone and its metabolites modulate 5HT1A and 5HT1B agonist effects on intermale aggression. Neurosci Biobehav Rev 23:325-336.

Sinha R, Garcia M, Paliwal P, Kreek MJ, Rounsaville BJ (2006) Stress-induced cocaine craving and hypothalamic-pituitary-adrenal responses are predictive of cocaine relapse outcomes. Arch Gen Psychiatry 63:324-331.

Smiley JF, Levey AI, Ciliax BJ, Goldman-Rakic PS (1994) D1 dopamine receptor immunoreactivity in human and monkey cerebral cortex: predominant and extrasynaptic localization in dendritic spines. Proc Natl Acad Sci USA 91:5720-5724.

297

Smith RF, Medici CN, Raap DK (1999) Enduring behavioral effects of weaning-through-puberty cocaine dosing in the rat. Psychobiology 27:432-437.

Sobrian SK, Marr L, Ressman K (2003) Prenatal cocaine and/or nicotine exposure produces depression and anxiety in aging rats. Progress in Neuro-Psychopharmacology & Biological Psychiatry 27:501-518.

Sorg BA, Davidson DL, Kalivas PW, Prasad BM (1997) Repeated daily cocaine alters subsequent cocaine-induced increase of extracellular dopamine in the medial prefrontal cortex. J Pharmacol Exp Ther 281:54-61.

Spangler R, Zhou Y, Maggos CE, Zlobin A, Ho A, Kreek MJ (1996) Dopamine antagonist and "binge' cocaine effects on rat opioid and dopamine transporter mRNAs. Neuroreport 7:2196-2200.

Spear L (2000) The adolescent brain and age-related behavioral manifestations. Neurosci Biobehav Rev 24:417-463.

Spector S, Tarver J, Berkowitz B (1972) Effects of drugs and physiological factors in the disposition of catecholamines in blood vessels. Pharmacol Rev 24:191-202.

Starkey NJ, Normington G, Bridges NJ (2007) The effects of individual housing on 'anxious' behaviour in male and female gerbils. Physiol Behav 90:545-552.

Stefanski R, Ziolkowska B, Kusmider M, Mierzejewski P, Wyszogrodzka E, Kolomanska P, Dziedzicka-Wasylewska M, Przewlocki R, Kostowski W (2007) Active versus passive cocaine administration: differences in the neuroadaptive changes in the brain dopaminergic system. Brain Res 1157:1-10.

Summers CH, Korzan WJ, Lukkes JL, Watt MJ, Forster GL, Overli O, Hoglund E, Larson ET, Ronan PJ, Matter JM, Summers TR, Renner KJ, Greenberg N (2005) Does serotonin influence aggression? comparing regional activity before and during social interaction. Physiol Biochem Zool 78:679-694.

Suthanthirarajan N, Subrahmanyam S (1983) Influence of isolation stress on monoamine levels in discrete regions of the rat brain correlated with behaviour. Indian J Physiol Pharmacol 27:101-108.

Suzuki M, Hurd YL, Sokoloff P, Schwartz JC, Sedvall G (1998) D3 dopamine receptor mRNA is widely expressed in the human brain. Brain Res 779:58-74.

Svensson P, Hurd YL (1998) Specific reductions of striatal prodynorphin and D1 dopamine receptor messenger RNAs during cocaine abstinence. Brain Res Mol Brain Res 56:162-168.

Teoh SK, Sarnyai Z, Mendelson JH, Mello NK, Springer SA, Sholar JW, Wapler M, Kuehnle JC, Gelles H (1994) Cocaine effects on pulsatile secretion of ACTH in men. J Pharmacol Exp Ther 270:1134-1138.

Ter Horst GJ, Wichmann R, Gerrits M, Westenbroek C, Lin Y (2009) Sex differences in stress responses: focus on ovarian hormones. Physiol Behav 97:239-249.

Terranova JP, Perio A, Worms P, Le Fur G, Soubrie P (1994) Social olfatory recognition in rodents: deterioration with age, cerebral ischaemia and septal lesion. Behav Pharmacol 5:90-98.

Tobin H, Logue AW (1994) Self-control across species (Columba livia, Homo sapiens, and Rattus norvegicus). J Comp Psychol 108:126-133.

Tuinier S, Verhoeven WM, van Praag HM (1995) Cerebrospinal fluid 5-hydroxyindolacetic acid and aggression: a critical reappraisal of the clinical data. Int Clin Psychopharmacol 10:147-156.

Ungless MA, Whistler JL, Malenka RC, Bonci A (2001) Single cocaine exposure in vivo induces long-term potentiation in dopamine neurons. Nature 411:583-587.

Van de Kar LD, Javed A, Zhang Y, Serres F, Raap DK, Gray TS (2001) 5-HT2A receptors stimulate ACTH, corticosterone, oxytocin, renin, and prolactin release and activate hypothalamic CRF and oxytocin-expressing cells. J Neurosci 21:3572-3579.

298

van der Vegt BJ, Lieuwes N, Cremers TI, de Boer SF, Koolhaas JM (2003a) Cerebrospinal fluid monoamine and metabolite concentrations and aggression in rats. Horm Behav 44:199-208.

van der Vegt BJ, Lieuwes N, van de Wall EH, Kato K, Moya-Albiol L, Martinez-Sanchis S, de Boer SF, Koolhaas JM (2003b) Activation of serotonergic neurotransmission during the performance of aggressive behavior in rats. Behav Neurosci 117:667-674.

van Erp AM, Miczek KA (2000) Aggressive behavior, increased accumbal dopamine, and decreased cortical serotonin in rats. J Neurosci 20:9320-9325.

Van Oortmerssen GA, Dijk DJ, Schuurman T (1987) Studies in wild house mice. II. Testosterone and aggression. Horm Behav 21:139-152.

Vanderschuren LJ, Kalivas PW (2000) Alterations in dopaminergic and glutamatergic transmission in the induction and expression of behavioral sensitization: a critical review of preclinical studies. Psychopharmacology (Berl) 151:99-120.

Varlinskaya EI, Spear LP, Spear NE (1999) Social behavior and social motivation in adolescent rats: role of housing conditions and partner's activity. Physiol Behav 67:475-482.

Veenema AH, Neumann ID (2007) Neurobiological mechanisms of aggression and stress coping: a comparative study in mouse and rat selection lines. Brain Behav Evol 70:274-285.

Veening JG, Coolen LM, de Jong TR, Joosten HW, de Boer SF, Koolhaas JM, Olivier B (2005) Do similar neural systems subserve aggressive and sexual behaviour in male rats? Insights from c-Fos and pharmacological studies. Eur J Pharmacol 526:226-239.

Vergnes M, Depaulis A, Boehrer A, Kempf E (1988) Selective increase of offensive behavior in the rat following intrahypothalamic 5,7-DHT-induced serotonin depletion. Behav Brain Res 29:85-91.

Vescovi PP, Coiro V, Volpi R, Passeri M (1992) Diurnal variations in plasma ACTH, cortisol and beta-endorphin levels in cocaine addicts. Horm Res 37:221-224.

Volkow ND, Fowler JS, Wang GJ, Hitzemann R, Logan J, Schlyer DJ, Dewey SL, Wolf AP (1993) Decreased dopamine D2 receptor availability is associated with reduced frontal metabolism in cocaine abusers. Synapse 14:169-177.

Volkow ND, Wang GJ, Fowler JS, Logan J, Gatley SJ, Gifford A, Hitzemann R, Ding YS, Pappas N (1999) Prediction of reinforcing responses to psychostimulants in humans by brain dopamine D2 receptor levels. Am J Psychiatry 156:1440-1443.

Volkow ND, Chang L, Wang GJ, Fowler JS, Ding YS, Sedler M, Logan J, Franceschi D, Gatley J, Hitzemann R, Gifford A, Wong C, Pappas N (2001) Low level of brain dopamine D2 receptors in methamphetamine abusers: association with metabolism in the orbitofrontal cortex. Am J Psychiatry 158:2015-2021.

Walker QD, Francis R, Cabassa J, Kuhn CM (2001) Effect of ovarian hormones and estrous cycle on stimulation of the hypothalamo-pituitary-adrenal axis by cocaine. J Pharmacol Exp Ther 297:291-298.

Weddington WW, Brown BS, Haertzen CA, Cone EJ, Dax EM, Herning RI, Michaelson BS (1990) Changes in mood, craving, and sleep during short-term abstinence reported by male cocaine addicts. A controlled, residential study. Arch Gen Psychiatry 47:861-868.

White SM, Kucharik RF, Moyer JA (1991) Effects of serotonergic agents on isolation-induced aggression. Pharmacol Biochem Behav 39:729-736.

Wichniak A, Brunner H, Ising M, Pedrosa Gil F, Holsboer F, Friess E (2004) Impaired hypothalamic-pituitary-adrenocortical (HPA) system is related to severity of benzodiazepine withdrawal in patients with depression. Psychoneuroendocrinology 29:1101-1108.

Williams JM, Steketee JD (2005) Time-dependent effects of repeated cocaine administration on dopamine transmission in the medial prefrontal cortex. Neuropharmacology 48:51-61.

Wilson JM, Nobrega JN, Carroll ME, Niznik HB, Shannak K, Lac ST, Pristupa ZB, Dixon LM, Kish SJ (1994) Heterogeneous subregional binding patterns of 3H-WIN 35,428 and 3H-GBR

299

12,935 are differentially regulated by chronic cocaine self-administration. J Neurosci 14:2966-2979.

Winstanley CA, Theobald DE, Cardinal RN, Robbins TW (2004) Contrasting roles of basolateral amygdala and orbitofrontal cortex in impulsive choice. J Neurosci 24:4718-4722.

Wise RA (2002) Brain reward circuitry: insights from unsensed incentives. Neuron 36:229-240.

Wongwitdecha N, Marsden CA (1996) Social isolation increases aggressive behaviour and alters the effects of diazepam in the rat social interaction test. Behav Brain Res 75:27-32.

Wood RD, Spear LP (1998) Prenatal cocaine alters social competition of infant, adolescent, and adult rats. Behav Neurosci 112:419-431.

Xia Y, Goebel DJ, Kapatos G, Bannon MJ (1992) Quantitation of rat dopamine transporter mRNA: effects of cocaine treatment and withdrawal. J Neurochem 59:1179-1182.

Yamamuro Y (2006) Social behavior in laboratory rats: Applications for psycho-neuroethology studies. Animal Science Journal 77:386-394.

Zangrossi H, Jr., Viana MB, Graeff FG (1999) Anxiolytic effect of intra-amygdala injection of midazolam and 8-hydroxy-2-(di-n-propylamino)tetralin in the elevated T-maze. Eur J Pharmacol 369:267-270.

Zhou Y, Spangler R, Schlussman S, Ho A, Kreek M (2003) Alterations in hypothalamic-pituitary-adrenal axis activity and in levels of proopiomelanocortin and corticotropin-releasing hormone-receptor 1 mRNAs in the pituitary and hypothalamus of the rat during chronic 'binge' cocaine and withdrawal. Brain Res 964:187-199.

Zhou Y, Spangler R, LaForge KS, Maggos CE, Ho A, Kreek MJ (1996a) Modulation of CRF-R1 mRNA in rat anterior pituitary by dexamethasone: correlation with POMC mRNA. Peptides 17:435-441.

Zhou Y, Spangler R, LaForge KS, Maggos CE, Ho A, Kreek MJ (1996b) Corticotropin-releasing factor and type 1 corticotropin-releasing factor receptor messenger RNAs in rat brain and pituitary during "binge"-pattern cocaine administration and chronic withdrawal. J Pharmacol Exp Ther 279:351-358.

300

Chapter 5

General Conclusion

5. General Conclusion

A global analysis of the data reported in the present work allows to conclude that

the chronic exposure to cocaine throughout adolescence produced sex-dependent

hormonal, neurochemical and behavioural short and long-term alterations.

Global alterations in adolescent male rats chronically exposed to cocaine

In male rats, the HPA axis was both activated by acute cocaine administration after

chronic cocaine treatment, and affected in a long-term perspective. Although the basal

activity was not affected, a disrupted HPA axis response under anxiogenic conditions was

observed in male rats 10 days after withdrawal, as indicated by the presence of higher

corticosterone levels in response to the EPM.

Chronic cocaine exposure throughout adolescence also led to short and long-

lasting disruptive effects on the HPG axis function in male rats, as indicated by the

decreased testosterone levels observed both 30 minutes and 10 days after the last

administration of cocaine. Concurrently these animals also showed decreased relative

gonadal weights.

Within the dopaminergic and serotonergic neurotransmission systems, an overall

tolerance to the acute effects of cocaine was observed after chronic cocaine

administration. However, the neurochemical data obtained 1 day after the last

administration of cocaine, revealed increased dopaminergic activity, which led us to

suggest that the expected decrease in dopaminergic activity could have been masked by

exposure to cocaine cues-induced increases in dopaminergic activity, since the animals

were sacrificed in the same room where cocaine was daily administered. This may

indicate a high susceptibility of adolescent rats to environmental cues, with male rats

showing a greater involvement of the nigrostriatal pathway in this process.

In a long-term perspective, the dopaminergic system in male rats was undoubtedly

affected by the cocaine treatment, as demonstrated by increased dopaminergic activity in

the nigrostriatal pathway, and supported by increased expression levels of TH mRNA in

the SN-VTA. This process may represent an attempt to compensate for a chronic cocaine-

induced depression of the dopaminergic system.

Long-lasting effects of cocaine treatment were also found in the serotonergic

activity in male rats. Increased 5-HT levels in SN-VTA were observed in cocaine-treated

males, and as suggested for the dopaminergic activity, such serotonergic activity increase

303

may represent an attempt to counteract the downregulation of this system during

withdrawal.

The behavioural studies performed seem to indicate that cocaine exposure during

adolescence interfered with the adaptive responses to environmental challenges.

Impaired risk-assessment during the EPM apparatus exploration was observed in

male rats, both at short- and long-term withdrawal. Paralleling impaired risk-assessment

during short-term withdrawal, our data showed impaired dopaminergic and serotonergic

neurotransmission in the prefrontal cortex and impaired dopaminergic neurotransmission

in the amygdala. Supporting the altered risk-assessment during long-term withdrawal it

was observed impaired dopaminergic and serotonergic responses in the amygdala. These

neurochemical alterations were probably related with the failure of cocaine-treated males

to exhibit an adequate behaviour when exploring the EPM. Importantly, cocaine-induced

impaired risk-assessment presents as an important potential cause for the high rates of

drug consumption prevalence during adolescence, and with increased vulnerability to

relapse during withdrawal.

Cocaine treatment also intensified social investigation and reduced defensive

behaviour during the social conflict provided by the resident-intruder paradigm, indicating

that the cocaine treatment affected the ability of male rats to properly interact with

conspecifics. Moreover, the reduced fear presented by the cocaine-treated animals, as

indicated by the decrease in flight behaviour, suggests altered risk-assessment properties,

supporting the results obtained in the EPM. Likewise, the neurochemical data obtained

after the resident-intruder paradigm, namely the altered function observed in prefrontal

cortex, amygdala and hippocampus, supports the alterations in the social behaviour

observed in these animals.

Global alterations in adolescent female rats chronically exposed to cocaine

In female rats, the hormonal data obtained showed no alterations in HPA axis

activity and effects of cocaine treatment on the HPG axis activity were also difficult to

infer.

However, like in males, also in female rats, an overall tolerance to the acute effects

of cocaine in the dopaminergic and serotonergic neurotransmission was observed after

chronic cocaine administration. Moreover, females also presented an increased

dopaminergic activity 1 day after withdrawal, suggesting, like in males, the presence of

304

high susceptibility to environmental cues. Interestingly, females presented a greater

involvement of amygdala in this process.

Cocaine-treatment also induced long-term effects on dopaminergic and

serotonergic neurotransmission in female rats, as indicated by increased dopaminergic

activity in the amygdala and hippocampus, and increased serotonergic activity in the

hippocampus and Nac observed throughout long-term withdrawal. Among other effects,

these long-term alterations could contribute to an increased vulnerability to relapse and

interfere with processes of learning and memory formation.

Diverging from males, in female rats, no behavioural differences were observed

between cocaine-treated and controls when performing the EPM.

In conclusion, the results obtained in the present study indicate the presence of

sex-differences in the short- and long-term effects of cocaine treatment during

adolescence. Male rats were more intensively affected than females. However, certain

conditions of the experimental procedure employed in the present study may have

contributed to mask the effects of cocaine in the female rats. Namely, the social isolation

that animals were submitted to, may have contributed to this effect. Sex-differences in the

susceptibility to the effects of stress are known, and several findings indicate an increased

vulnerability of females in this field. Hence, social isolation may have masked more

prominently the cocaine exposure effects in female rats.

Although cocaine exposure effects during adulthood were not evaluated in the

present study, compared to published data, our results seem indicate less differences in

dopaminergic and serotonergic neurotransmission between saline and cocaine-treated

animals. Namely, adolescent rats treated chronically with cocaine showed few acute

effects as compared with data from studies carried out during the adulthood. This may

indicate an improved adaptive response, which may be a consequence of the increased

plasticity that characterizes the adolescent CNS maturation process. On the other hand,

our results also seem to indicate low levels of anxiety-like, depressive-like and aggression

behaviours during withdrawal from cocaine exposure during adolescence as compared

with data from literature on the effects of cocaine in adult animals. In fact, provides further

evidence in supporting previous findings suggesting that adolescents respond differently

from older animals to psychostimulants.

This age related effect may be involved in the increased vulnerability to drug use

during adolescence. Indeed, the impairment in risk assessment observed in male rats

305

exposed to cocaine during adolescence may lead to a compromise decision making

involving choices of high risks and low benefits. Moreover, this could represent an

important risk factor for the increased susceptibility to the maintenance of drug use and for

the increased risk of relapse after a drug-free period.

From a social perspective, data obtained in this study presents an increased

relevance for the understanding of the relationship between adolescents and drug use

and can be issued to better structure drug abuse prevention in the adolescence.

306

Appendix 1

Article

“Hormonal, neurochemical, and behavioral response to a Forced Swim Test in

adolescent rats throughout cocaine withdrawal”

Hormonal, Neurochemical, and BehavioralResponse to a Forced Swim Test in

Adolescent Rats throughoutCocaine Withdrawal

Cecılia Juliana Alves,a,b Ana Magalhaes,a

Teresa Summavielle,a,c Pedro Melo,a Liliana De Sousa,a,b

Maria Amelia Tavares,a,d and Pedro R.R. Monteiroa,c

aIBMC–Instituto de Biologia Molecular e Celular, Universidade do Porto,Porto, Portugal

bInstitute of Biomedical Sciences Abel Salazar, University of Porto, Porto, PortugalcEscola Superior de Tecnologia da Saude do Porto do IPP, Porto, Portugal

dInstitute of Anatomy, Medical School of the University of Porto, Porto, Portugal

The use of cocaine in adults has been linked to depression and/or anxiety. Severalstudies have shown an association between cocaine-primed craving and depressivesymptoms. In animal models, the forced swim test (FST) is frequently used for screen-ing depressive-like behavior. This study aimed to verify the presence of depression-likesymptoms in adolescent rats after chronic cocaine exposure by analyzing behavior ina FST. The subsequent alterations in neurotransmitters and hypothalamus-pituitary-adrenal axis activity induced by this test were also analyzed. Both male and femaleadolescent Wistar rats were submitted to a chronic “binge” pattern of administrationof cocaine hydrochloride, and subjects were tested in a forced swim test 2 days aftercocaine’s last administration. At the end of the behavioral test, trunk blood was col-lected for quantification of corticosterone plasma levels, and hypothalamus, prefrontalcortex, amygdala, and hippocampus were dissected for neurochemical determinations.No significant differences were found in the behavior on the FST of both males andfemales after withdrawal from chronic cocaine administration. Nevertheless, plasmalevels of corticosterone were increased in cocaine-treated males, although not signifi-cantly (P = 0.065). In females cocaine failed to affect corticosterone levels. Of interest,neurochemical analyses showed that dopamine turnover was decreased in amygdalain cocaine-treated males (not significantly, P = 0.055). No significant differences werefound on neurotransmitter levels in the other brain regions analyzed. Withdrawal fromchronic cocaine administration during adolescence did not have a significant effecton stress-induced behavioral alterations, although the neurochemical response to thestressful situation provided by FTS seemed to be affected.

Key words: cocaine; chronic exposure; withdrawal; adolescence; rat; forced swim test;behavior; monoamines; corticosterone

Address for correspondence: Cecılia Juliana Alves, Unidade de Neuro-comportamento, IBMC–Instituto de Biologia Molecular e Celular, Ruado Campo Alegres, 823, 4150-180 Porto, Portugal. Voice: +351-226-074-900; fax: +351-226-099-157. [email protected]

Introduction

Several studies have shown that cocaineabuse and withdrawal has been linkedto the development of depressive and/oranxiety-like symptoms.1,2 In humans and rats,

Ann. N.Y. Acad. Sci. 1139: 366–373 (2008). C© 2008 New York Academy of Sciences.doi: 10.1196/annals.1432.047 366

Alves et al.: Cocaine Withdrawal and Stress Response 367

antidepressants can attenuate symptoms of co-caine withdrawal,2,3 confirming that depres-sion may be involved.4 Moreover, severe anxi-ety and depression provide part of the negativereinforcement associated with cocaine depen-dence and are an important motivational factorfor relapse and maintenance of repetitive cyclesof cocaine abuse.2

The forced swim test (FST) was proposed byPorsolt et al.5 as a model of depression and itis still used to screen antidepressants in rats.6,7

The FST is also often employed as a measure offear or emotionality in response to a stressor.8,9

In fact, this test is a potent psychophysiologi-cal stressor that is accompanied by a variety ofneurochemical, endocrine, and immunologicchanges.10,11 Alterations in the serotoninergicsystem induced by exposure to the FST, namely,an increase in the serotonin (5-HT) turnover inseveral brain areas has been reported.12 Thetest is also recognized to increase the activityof the hypothalamus-pituitary-adrenal (HPA)axis.13

Cocaine also seems to alter the behavioralresponse to stressful situations. Repeated co-caine decreases the avoidance response to anovel stimulus in rats,14 and in the FST post-natal exposure to cocaine increases immobilityin juvenile rats.15 On the other hand, stress isknown to contribute to reinstatement of drugabuse.16,17

Although adolescents represent a risk groupregarding drug abuse and addiction, experi-mental research evaluating the effects of psy-chostimulants has been mostly focused on theassessment of adult animals. In fact, previousstudies have shown that most illicit drug usebegins in adolescence,18,19 and that adolescentsare more likely to progress from use to depen-dence than adults.20

Adolescence is an ontogenetic transitionfrom the dependence of youth to the indepen-dence of the adulthood, and during this phasethe brain and the hormonal systems are still un-dergoing many complex changes.21 Disruptionof the development of the central nervous sys-tem at this stage may increase the probability

of developing psychopathologies such as drugdependence (reviewed by Spear21). Therefore,there is a growing interest in assessing the influ-ence that drug use can have on the developingadolescent.

The present study aims to verify the presenceof depressive symptoms in male and femaleadolescent rats after withdrawal from chroniccocaine administration, assessing the behaviorin a FST. The effects of cocaine treatment onthe subsequent alterations on brain neurotrans-mitters and in hypothalamus-pituitary-adrenalaxis activity induced by the FST were alsoanalyzed.

Material and Methods

Animal Model

Wistar rats born from nulliparous femalespurchased from Charles River LaboratoriesEspana S.A. (Barcelona, Spain) were used inthis experiment. Animals were maintained un-der a 12-h light/dark cycle in a temperature-and humidity-controlled room and given ad li-

bitum access to food and water. Institutionalguidelines regarding animal experimentationwere followed. All procedures used were ap-proved by the Portuguese Agency for An-imal Welfare (General Board of VeterinaryMedicine in compliance with the InstitutionalGuidelines and the European Convention).

On the day after birth, postnatal day 1 (PND1), litters were standardized, whenever possible,to 10 sex-balanced pups. Animals were weanedon PND 21 and housed individually on PND30 to avoid hormonal co-regulation. In orderto minimize the effects of social isolation, bothcontrol and treated animals were housed indi-vidually handled daily and were given cylindri-cal plastic tubes and soft paper for nest con-struction. One male and one female from eightlitters received intraperitoneal (i.p.) injections ofcocaine (3 × 15 mg/kg) from PND 35 to PND50, following a “binge” pattern regime: threetimes daily at hourly intervals, between 9 AM

368 Annals of the New York Academy of Sciences

and 11 AM. Controls were given isovolumetricsaline (0.9%) following the same experimentalprotocol. Cocaine was obtained from SigmaChemical Company (St. Louis, MO, USA).

Forced Swim Test

The FST procedure used was similar to theone described by Porsolt et al.5 Two days aftercocaine’s last administration, rats were placedinto a glass cylinder (19 cm in diameter and40 cm deep) of 22 ± 1 ◦C water filled to a depthof 30 cm for 15 min of pretest. Twenty-fourhours later, rats were again placed into the wa-ter for 5 min. Activity during the second swimtest was video-recorded for subsequent behav-ioral analysis. Videotapes were scored by anobserver using the Noldus Observer software(Observer 4.1 Noldus Information Technology,Wageningen, the Netherlands) and the follow-ing behavior categories were analyzed: immo-bility, struggling, diving, as well as right andleft rotation. The data were collected for fre-quency, duration, and latency. Immobility wasdefined as lack of movement of three paws andonly minimal movement of the fourth, or lackof movement of all four paws.22 Struggling wasdefined as vigorous movement of all four paws,with the rat in a relatively upright position.23

Blood and Tissue Collection

At the end of the behavioral session animalswere decapitated and the brain was rapidlyremoved. Amygdala, hippocampus, prefrontalcortex, and suprachiasmatic, pre-optic and par-aventricular nuclei of hypothalamus were dis-sected on ice (according to the atlas by Paxi-nos and Watson24), frozen in 2-methylbutanecooled over dry ice, and stored at −70 ◦C un-til used for neurotransmitter determinations.Trunk blood was collected in lithium heparin-coated tubes and centrifuged at 1600 × g for15 min, and the plasma obtained was kept at−70 ◦C until corticosterone (CORT) hormoneassay.

Hormone Assays

CORT plasma levels were measured byenzyme immunoassay, using a commerciallyavailable kit provided by IDS, Ltd. (Boldon,UK). The intra- and interassay coefficientsof variation were less than 6.6% and 8.6%,respectively.

Neurotransmitter Determinations

Concentrations of dopamine (DA) andserotonin (5-HT) and their metabolites, 5-hydroxyindole-3-acetic acid (5-HIAA), 3,4-dihydroxyphenylacetic acid (DOPAC), andhomovanillic acid (HVA) from the brain ar-eas collected were quantified by a modifiedmethod25 of high-performance liquid chro-matography with electrochemical detection(HPLC/EC) in a Gilson Medical Electronicssystem (Gilson, Middleton, WI, USA). The an-alytic column was a Supelco, Inc. (Bellefonte,PA, USA) Supelcosil LC-18 of 3 μM (7.5 cm ×4.6 mm). Concentrations of neurotransmitterswere calculated using standard curves gener-ated using standard amine purchased from theSigma Chemical Co. Neurotransmitter con-centrations were expressed in ng/mg protein.Protein content was determined by the Bio-Rad protein assay (Munchen, Germany), usingbovine serum albumin as a standard. The ratioof DOPAC and HVA to DA was determined foreach animal and used as an index of dopamineturnover rate; likewise, the ratio of 5-HIAA to5-HT was also determined and used as an in-dex of serotonin turnover rate.

Statistical Analyses

FST behavior during chronic cocaine with-drawal was analyzed by two-way ANOVAwith treatment (cocaine or saline) and sex asbetween-subject fixed-factors and behavioralmeasurements as the dependent variable. Thet-test was used as a post hoc test to analyze sta-tistically significant differences. For hormonaland neurochemical data, statistical significance

Alves et al.: Cocaine Withdrawal and Stress Response 369

between groups was assessed using the non-parametric Mann–Whitney U Test. The sta-tistical level of significance was considered atP < 0.05. All data are expressed as mean ±SEM. Statistical analyses were performed us-ing SPSS 14.0 software (SPSS INC. Chicago,Illinois, USA).

Results

Behavioral Data

Statistic analyses revealed an interaction be-tween sex and treatment for the latency to strug-gling behavior [F(1.28) = 4.969; P < 0.05]. Post

hoc comparisons using the t-test indicate that inthe control group the latency to struggling waslower in males [t(14) =−2.293 (two-tailed), P <

0.05]. No significant differences were found onthe behavior in the FST of both males and fe-males after withdrawal from chronic cocaineadministration (Table 1).

Hormonal Data

Analysis of hormonal data revealed thatwithin the male group, plasma levels of corti-costerone were increased after FST in cocaine-treated animals, although not significantly [Z =−1.848 (two-tailed), P = 0.065] (Fig. 1). Infemales the treatment did not affect CORTlevels.

Neurochemical Data

In the amygdala, within the control group, fe-males displayed increased levels of 5-HT, whencompared with those of control males, that al-most reached significance [Z = −1.922 (two-tailed) P = 0.055] (Fig. 2A) and also a lowerratio 5-HIAA/5-HT [Z = −2.242 (two-tailed)P < 0.05] (Fig. 2B). Also in this brain re-gion, cocaine decreased (although not signif-icantly) the ratio DOPAC/DA in male [Z =−1.922 (two-tailed), P = 0.055], but not in fe-male rats (Fig. 2C). Analysis of neurochemicaldata from prefrontal cortex, hippocampus, and TA

BLE

1.

Effe

cts

ofW

ithdr

awal

from

Coc

aine

Expo

sure

duri

ngA

dole

scen

ceon

Beha

vior

ina

FST

inM

ale

and

inFe

mal

eRa

ts

Stru

gglin

gIm

mob

ility

Div

ing

Rig

htro

tatio

nL

eftr

otat

ion

Freq

uenc

yM

ale

Salin

e(n

=9)

10

0.44

±0.3

428

.67

±2.

4836

.56

±2.

91C

ocai

ne(n

=9)

10

1.22

±0.

1031

.22

±2.

5332

.44

±3.

22Fe

mal

eSa

line

(n=

9)1

00.

44±

0.34

34.4

4.60

33.7

4.02

Coc

aine

(n=

8)1

00.

0.5

35.3

3.08

29.8

3.95

Dur

atio

n(se

c)M

ale

Salin

e(n

=9)

47.7

14.5

2–

0.37

±0.

36–

–C

ocai

ne(n

=9)

27.8

8.40

–1.

53±

1.29

––

Fem

ale

Salin

e(n

=9)

37.6

7.53

–0.

81±

0.54

––

Coc

aine

(n=

8)38

.23

±8.

73–

0.66

±0.

68–

–L

aten

cy(se

c)M

ale

Salin

e(n

=9)

0.88

±0.

33a

–25

3.40

±30

.91

4.85

±2.

482.

13±

0.93

Coc

aine

(n=

9)1.

28±

0.53

–24

7.17

±32

.91

3.36

±0.

713.

36±

1.49

Fem

ale

Salin

e(n

=9)

2.09

±0.

67a

–25

2.66

±31

.87

4.12

±1.

574.

81±

1.56

Coc

aine

(n=

8)5.

18±

2.43

–26

9.74

±30

.27

1.57

±0.

623.

75±

1.83

Not

e:D

ata

are

expr

esse

das

the

mea

nva

lue±

SEM

.Val

ues

inth

esa

me

colu

mn

with

the

sam

ele

tter

are

sign

ifica

ntly

diffe

rent

.a P

<0.

05.

370 Annals of the New York Academy of Sciences

Figure 1. Effects of withdrawal from cocaine ex-posure during adolescence on plasma levels of corti-costerone induced by forced swim test, both in maleand female rats. Each column represents the mean ±S.E.M for 6 animals per group. CORT, corticosterone.

hypothalamus after FST showed no differencesin levels of DA or 5-HT and their metabolitesor in turnover rates.

Discussion

Previous work in adult animals hasshown that withdrawal from cocaine inducesdepressive-like behavior.2,3 However, our re-sults did not show any significant effect ofcocaine withdrawal during adolescence on thebehavior of male and female rats on the FST.Increased passive behavioral responses in theFST such as immobility and decreased ac-tive behaviors like swimming or struggling arethought to be a clear indication of depressive-like symptoms,6,26 but in our study these mea-sures were not affected by cocaine treatment.

Following the criteria established to defineimmobility, we were not able to observe alteredimmobility behavior in any studied group ofanimals. Some studies have shown that therewas an age-dependent difference in the amountof time spent in immobility in a forced swimtest,8,27 with juveniles showing very little ev-idence of immobility in contrast to adults.23

However, at least one previous study showedthat the immobility response was present inadolescent rats.28 This absence of immobilitybehavior observed in our study could be due tomethodologic differences in experimental pro-cedures, such as apparatus dimensions, watertemperature, and/or criteria for defining im-

Figure 2. Effects of withdrawal from cocaine ex-posure during adolescence on the levels (ng/mgprotein) of 5-HT (A), on 5-HIAA/5-HT (B), and onDOPAC/DA ratio (C) in amygdala after a forcedswim test both in male and female rats. Each columnrepresents the mean ± SEM for 6 animals per group.DA, dopamine; DOPAC, 3,4-dihydroxyphenylaceticacid; 5-HT, serotonin; 5-HIAA, 5-hydroxyindolaceticacid. Columns with the same letter are significantlydifferent: a = P < 0.05.

mobility.29 However, it should be noted thatthere were no treatment effects in the timespent struggling either in males and females,which allow us to propose that the behavioralstrategy adopted to cope with the stressful con-dition provide by the FST was not affected bythe cocaine treatment and withdrawal. This ab-sence of significant differences between controland cocaine-exposed rats could be related tothe reduced behavioral and physiological re-sponse to stressful situations in rodents dur-ing the adolescent period indicated by somestudies.30,31 Moreover, cocaine was reported todisplay lower behavioral effects in adolescence

Alves et al.: Cocaine Withdrawal and Stress Response 371

than in adult rats.32 In addition, our experi-mental design requires animals to be housedindividually, in order to avoid variable socialinfluences that could normalize hormonal ef-fects, masking the possible effects of cocaineexposure.33 However, housing isolation itself,despite our efforts to minimize it through im-proved handling, may contribute to the devel-opment of depressive behavior,34 which couldinterfere with the possible effects of cocainetreatment regarding depressive-like behavioraldevelopment.

Beside the evaluation of the behavioral re-sponse to FST, alterations in physiological re-sponses to the stressful situation provided by theFST during withdrawal from chronic cocaineadministration were also analyzed. Althoughplasma levels of CORT were increased afterFST in cocaine-treated males, this increase wasnot significantly different. Thus, the hormonaldata from both male and female adolescentrats seems to indicate that the plasma levelsof CORT after performing the FST were notinfluenced by the withdrawal from chronic co-caine administration. Other authors have alsoshown a lack of cocaine effects on HPA re-sponse to stress. Seven days of exposure to15 mg/kg of cocaine failed to alter the CORTresponse to 20 min of immobilization 42 h af-ter the final injection,35 and no changes weredemonstrated in the CORT response to 60 minof immobilization 21 h after the final injectionof a 3- or 6-week “binge” cocaine regimen.36

However, Mantsch et al. reported a CORT re-sponse to a stressor, 30 min of restraint, duringacute (24-h) withdrawal from chronic cocaineadministration.37 In our study, the absence ofdifferences in CORT response to FST could berelated with the elevated basal levels of CORTpresent in adolescents. The fact that adoles-cents usually display higher CORT levels atbaseline than those of adults could contributeto a reduced percentage of increase when com-pared to adults.38,39 Moreover, age-related dis-continuities in the response of the HPA toboth stress and psychostimulants have been

demonstrated.30 The HPA axis seems to be hy-poresponsive to external perturbations duringadolescence.39 Since both saline and cocaine-treated rats showed a similar CORT responseto the FST, it seems that chronic cocaine ad-ministration did not result in the developmentof tolerance or sensitization of the HPA axisresponse to a novel stressor.

The neurochemical data presented indicatethat within the control group there was an in-crease in the 5-HT turnover in the amygdalain the adolescent males. This increase is con-sistent with previous reports that examined theeffects of other stressors on the 5-HT turnoverin the brain.40,41 Besides, such an effect hasalready been demonstrated in the amygdalaof adult male rats as a result of FST expo-sure.12 Data presented here reveal a sex ef-fect in the serotonergic activity of control ani-mals that was not observed in cocaine-exposedrats, which may be a consequence of cocainewithdrawal. Moreover, although not signifi-cantly, withdrawal from chronic cocaine de-creases the ratio DOPAC/DA in male rats. To-gether, these results seem to show that cocainetreatment during the adolescence did affectthe neurotransmitter system’s response to theFST.

In conclusion, although withdrawal fromchronic cocaine administration during adoles-cence seems not to have had a significant effecton stress-induced behavioral alterations, neu-rochemical data indicate that the response tothe stressful situation created by the FTS wasaffected. These results are substantially differ-ent from the data obtained in studies carriedout in adult rats, which do show stronger ef-fects.38,42 Thus, the data provided by this workmay confirm previous findings suggesting thatadolescents respond differently from older ani-mals to both stress and psychostimulants.39,43,44

This age-related effect could be one of thereasons for the elevated prevalence of druguse and dependence during adolescence.38 In-deed, age-related discontinuities in response topsychological stress have been indicated as a

372 Annals of the New York Academy of Sciences

psychobiological risk for increased vulnerabil-ity to drug abuse.45,46

Acknowledgments

This work was supported by Fundacaopara a Ciencia e Tecnologia (FCT) ProjectPOCTI/ESP/43630/1999 and Programade Financiamento Plurianual (IBMC).Cecılia Juliana Alves, Ana Magalhaes,and Teresa Summavielle were supportedby FCT Grants SFRH/BD/17195/2004,SFRH/BPD/19200/2004, and SFRH/BPD/20997/2004, respectively.

Conflicts of Interest

The authors declare no conflicts of interest.

References

1. Gawin, F.H. 1991. Cocaine addiction: psychologyand neurophysiology. Science 251: 1580–1586.

2. Markou, A., R.L. Hauger & G.F. Koob. 1992.Desmethylimipramine attenuates cocaine with-drawal in rats. Psychopharmacology (Berl.) 109: 305–314.

3. Gawin, F.H. et al. 1989. Desipramine facilitation ofinitial cocaine abstinence. Arch. Gen. Psychiatry 46:117–121.

4. Pliakas, A.M. et al. 2001. Altered responsiveness tococaine and increased immobility in the forced swimtest associated with elevated cAMP response element-binding protein expression in nucleus accumbens.J. Neurosci. 21: 7397–7403.

5. Porsolt, R.D., M. Le Pichon & M. Jalfre. 1977. De-pression: a new animal model sensitive to antidepres-sant treatments. Nature 266: 730–732.

6. Lopez-Rubalcava, C. & I. Lucki. 2000. Strain differ-ences in the behavioral effects of antidepressant drugsin the rat forced swimming test. Neuropsychopharmacol-

ogy 22: 191–199.7. Rogoz, Z. et al. 2005. Effects of combined treat-

ment with imipramine and metyrapone on the im-mobility time, the activity of hypothalamo-pituitary-adrenocortical axis and immunological parametersin the forced swimming test in the rat. J. Physiol. Phar-

macol. 56: 49–61.8. Bilitzke, P.J. & M.W. Church. 1992. Prenatal cocaine

and alcohol exposures affect rat behavior in a stress

test (the Porsolt swim test). Neurotoxicol. Teratol. 14:359–364.

9. Overstreet, D.H. et al. 2000. Enduring effects of pre-natal cocaine administration on emotional behaviorin rats. Physiol. Behav. 70: 149–156.

10. Abel, E.L. 1993. Physiological correlates of the forcedswim test in rats. Physiol. Behav. 54: 309–317.

11. Shu, J., J.R. Stevenson & X. Zhou. 1993. Modu-lation of cellular immune responses by cold waterswim stress in the rat. Dev. Comp. Immunol. 17: 357–371.

12. Connor, T.J., J.P. Kelly & B.E. Leonard. 1997. Forcedswim test-induced neurochemical endocrine, and im-mune changes in the rat. Pharmacol. Biochem. Behav. 58:961–967.

13. Connor, T.J. et al. 2000. Effect of subchronic antide-pressant treatments on behavioral, neurochemical,and endocrine changes in the forced-swim test. Phar-

macol. Biochem. Behav. 65: 591–597.14. Sorg, B. et al. 2002. Repeated cocaine decreases the

avoidance response to a novel aversive stimulus inrats. Psychopharmacology (Berl.) 163: 9–19.

15. Magalhaes, A., M.A. Tavares & L. De Sousa. 2002.Postnatal cocaine exposure: effects on behavior ofrats in forced swim test. Ann. N.Y. Acad. Sci. 965:529–534.

16. Erb, S., Y. Shaham & J. Stewart. 1996. Stress rein-states cocaine-seeking behavior after prolonged ex-tinction and a drug-free period. Psychopharmacology

(Berl.) 128: 408–412.17. Shaham, Y., S. Erb & J. Stewart. 2000. Stress-induced

relapse to heroin and cocaine seeking in rats: a review.Brain Res. Rev. 33: 13–33.

18. Estroff, T.W., R.H. Schwartz & N.G. Hoffmann.1989. Adolescent cocaine abuse- addictive potential,behavioral and psychiatric effects. Clin. Pediat. 28:550–555.

19. Chen, K. & D.B. Kandel. 1995. The natural historyof drug use from adolescence to the mid-thirties in ageneral population sample. Am. J. Public Health. 85:41–7.

20. Chen, K., D.B. Kandel & M. Davies. 1997. Relation-ships between frequency and quantity of marijuanause and last year proxy dependence among adoles-cents and adults in the United States. Drug Alcohol

Depend. 46: 53–67.21. Spear, L. 2000. The adolescent brain and age-related

behavioral manifestations. Neurosci. Biobehav. Rev. 24:417–463.

22. Viglinskaya, I.V. et al. 1995. To drink or not to drink:tests of anxiety and immobility in alcohol-preferringand alcohol-nonpreferring rat strains. Physiol. Behav.

57: 937–941.23. Hansen-Trench, L.S. & S. Barron. 2005. Effects of

neonatal alcohol and/or cocaine exposure on stress

Alves et al.: Cocaine Withdrawal and Stress Response 373

in juvenile and adult female rats. Neurotoxicol. Teratol.

27: 55–63.24. Paxinos, G. & C. Watson. 1998. The Rat Brain in Stereo-

taxic Coordinates, 4th ed. Academic Press. San Diego.25. Ali, S.F., S.N. David & G.D. Newport. 1993. Age-

related susceptibility to MPTP-induced neurotoxicityin mice. Neurotoxicology 14: 29–34.

26. Detke, M.J., M. Rickels & I. Lucki. 1995. Active be-haviors in the rat forced swimming test differentiallyproduced by serotonergic and noradrenergic antide-pressants. Psychopharmacology (Berl.) 121: 66–72.

27. Molina, V.A., J.M. Wagner & L.P. Spear. 1994. Thebehavioral response to stress is altered in adult ratsexposed prenatally to cocaine. Physiol. Behav. 55: 941–945.

28. Cannizzaro, C. et al. 2007. Perinatal exposure to 5-metoxytryptamine, behavioural-stress reactivity andfunctional response of 5-HT1A receptors in the ado-lescent rat. Behav Brain Res. 186: 98–106.

29. Abel, E.L. 1993. Rat offspring sired by males treatedwith alcohol. Alcohol 10: 237–242.

30. Laviola, G. et al. 2002. Peculiar response of adoles-cent mice to acute and chronic stress and to am-phetamine: evidence of sex differences. Behav. Brain

Res. 130: 117–125.31. Macri, S. et al. 2002. Risk taking during exploration of

a plus-maze is greater in adolescent than in juvenileor adult mice. Animal Behaviour 64: 541–546.

32. Collins, S. & S. Izenwasser. 2002. Cocaine differen-tially alters behavior and neurochemistry in periado-lescent versus adult rats. Brain Res. Dev. Brain Res. 138:27–34.

33. de Jong, I.E., M.S. Oitzl & E.R. de Kloet. 2007.Adrenalectomy prevents behavioural sensitisation ofmice to cocaine in a genotype-dependent manner.Behav. Brain Res. 177: 329–339.

34. Brenes Saenz, J.C., O.R. Villagra & J. FornagueraTrias. 2006. Factor analysis of Forced Swimming test,Sucrose Preference test and Open Field test on en-riched, social and isolated reared rats. Behav. Brain

Res. 169: 57–65.35. Levy, A.D. et al. 1994. Influence of repeated cocaine

exposure on the endocrine and behavioral-responsesto stress in rats. Psychopharmacology 113: 547–554.

36. Sarnyai, Z. et al. 1998. Neuroendocrine-related ef-fects of long-term, ‘binge’ cocaine administration:diminished individual differences in stress-inducedcorticosterone response. Neuroendocrinology 68: 334–344.

37. Mantsch, J.R. et al. 2007. Restraint-induced corti-costerone secretion and hypothalamic CRH mRNAexpression are augmented during acute withdrawalfrom chronic cocaine administration. Neurosci. Lett.

415: 269–273.38. Schramm-Sapyta, N.L., R.W. Morris & C.M. Kuhn.

2006. Adolescent rats are protected from the con-ditioned aversive properties of cocaine and lithiumchloride. Pharmacol. Biochem. Behav. 84: 344–352.

39. Adriani, W. & G. Laviola. 2000. A unique hormonaland behavioral hyporesponsivity to both forced nov-elty and d-amphetamine in periadolescent mice. Neu-

ropharmacology 39: 334–346.40. Dunn, A.J. 1988. Stress-related changes in cerebral

catecholamine and indoleamine metabolism: lack ofeffect of adrenalectomy and corticosterone. J. Neu-

rochem. 51: 406–412.41. Inoue, T., K. Tsuchiya & T. Koyama. 1994. Regional

changes in dopamine and serotonin activation withvarious intensity of physical and psychological stressin the rat brain. Pharmacol. Biochem. Behav. 49: 911–920.

42. O’Dell, L.E. et al. 2007. Adolescent nicotine expo-sure produces less effective measures of withdrawalrelative to adult nicotine exposure in male rats. Neu-

rotoxicol. Teratol. 29: 17–22.43. Infurna, R.N. & L.P. Spear. 1979. Developmen-

tal changes in amphetamine-induced taste aversions.Pharmacol. Biochem. Behav. 11: 31–35.

44. Stansfield, K.H. & C.L. Kirstein. 2005. Neurochem-ical effects of cocaine in adolescence compared toadulthood. Brain Res. Dev. Brain Res. 159: 119–125.

45. Adriani, W., F. Chiarotti & G. Laviola. 1998. Ele-vated novelty seeking and peculiar d-amphetaminesensitization in periadolescent mice compared withadult mice. Behav. Neurosci. 112: 1152–1166.

46. Laviola, G. et al. 1995. Cocaine sensitization in peri-adolescent and adult rats. J. Pharmacol. Exp. Ther. 275:345–357.