characterization of disposition of deltamethrin, cis

213
CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS-PERMETHRIN AND TRANS-PERMETHRIN AS A FUNCTION OF AGE IN SPRAGUE-DAWLEY RATS by MANOJ AMARANENI (Under the Direction of James V. Bruckner) ABSTRACT Although the blood and tissue partition coefficients (PCs) for pyrethroids in adult rats have previously been published, no one has evaluated the influence of maturation on the systemic disposition of pyrethroids in rats. This series of studies addressed this deficit by testing the hypothesis that the distribution of pyrethroids in immature rats will be significantly different from that in adults. Steady-state levels of deltamethrin (DLM), cis-permethrin (CIS) and trans-permethrin (TRANS) were obtained using subcutaneously implanted Alzet pumps ® in Sprague-Dawley (SD) rats. For all three compounds, fat exhibited far higher PC values than brain, liver and skeletal muscle in adult rats. Brain and liver exhibited the lowest PCs followed by skeletal muscle. For all three compounds, the tissue:plasma PCs in PND 15 pups were found to be significantly higher than in adult rats. Alternatively, there was no distinct pattern, in the differences in tissue:plasma PCs between PND 21 and adult rats. An in situ brain perfusion technique was utilized to selectively investigate the role of plasma binding, membrane transporters and the blood brain barrier (BBB) on the

Upload: others

Post on 14-Jan-2022

6 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS-PERMETHRIN

AND TRANS-PERMETHRIN AS A FUNCTION OF AGE IN SPRAGUE-DAWLEY

RATS

by

MANOJ AMARANENI

(Under the Direction of James V. Bruckner)

ABSTRACT

Although the blood and tissue partition coefficients (PCs) for pyrethroids in adult

rats have previously been published, no one has evaluated the influence of maturation

on the systemic disposition of pyrethroids in rats. This series of studies addressed this

deficit by testing the hypothesis that the distribution of pyrethroids in immature rats will

be significantly different from that in adults. Steady-state levels of deltamethrin (DLM),

cis-permethrin (CIS) and trans-permethrin (TRANS) were obtained using

subcutaneously implanted Alzet pumps® in Sprague-Dawley (SD) rats. For all three

compounds, fat exhibited far higher PC values than brain, liver and skeletal muscle in

adult rats. Brain and liver exhibited the lowest PCs followed by skeletal muscle. For all

three compounds, the tissue:plasma PCs in PND 15 pups were found to be significantly

higher than in adult rats. Alternatively, there was no distinct pattern, in the differences in

tissue:plasma PCs between PND 21 and adult rats.

An in situ brain perfusion technique was utilized to selectively investigate the role

of plasma binding, membrane transporters and the blood brain barrier (BBB) on the

Page 2: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

brain uptake of DLM. The data suggested that under normal physiological conditions,

plasma binding along with the BBB plays a protective role in restricting the entry of DLM

into the brain. At physiological albumin concentrations and low exposure levels, a

passive, non-saturable transport of DLM predominates at the BBB. Brain uptake of DLM

was determined as a function of age-dependent changes in plasma protein binding and

the BBB integrity. Adult brain uptake of DLM from pediatric and adult rat and human

plasmas did not vary significantly. When measured as a function of the BBB integrity,

uptake in PND 15 and PND 21 pups was significantly greater than in adults.

The data presented here demonstrate that the steady-state tissue:plasma PCs for

pyrethroids were inversely proportional to age. These differences become more

pronounced as the age diminishes. At low exposure levels of DLM, ontogenic

differences in plasma binding do not affect its brain uptake. However, increased brain

entry of DLM in younger rats results from increased permeability of the BBB in these

age groups.

INDEX WORDS: Pyrethroids, DLM, CIS, TRANS, tissue:plasma partition coefficients,

Blood Brain Barrier, in situ brain perfusion, membrane transporters.

Page 3: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS-PERMETHRIN

AND TRANS-PERMETHRIN AS A FUNCTION OF AGE IN SPRAGUE-DAWLEY

RATS

by

MANOJ AMARANENI

B.Pharmacy, Andhra University, India, 2010

A Dissertation Submitted to the Graduate Faculty of The University of Georgia in Partial

Fulfillment of the Requirements of the Degree

DOCTOR OF PHILOSOPHY

ATHENS, GEORGIA

2016

Page 4: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

© 2016

Manoj Amaraneni

All Rights Reserved

Page 5: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS-PERMETHRIN

AND TRANS-PERMETHRIN AS A FUNCTION OF AGE IN SPRAGUE-DAWLEY

RATS

By

MANOJ AMARANENI

Major Professor: James V Bruckner

Committee: Catherine A White

Brian S Cummings

Michael G Bartlett

Randall Tackett

Electronic Version Approved

Suzanne Barbour Dean of the Graduate School The University of Georgia December 2016

Page 6: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

iv

DEDICATION

I would like to dedicate my dissertation to my parents and my wife who gave me

strength and faith to succeed.

Page 7: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

v

ACKNOWLEDGEMENTS

I cordially thank my major advisor, Dr. James V. Bruckner, for his support,

encouragement and guidance during my graduate years. His guidance helped me in all

the time of research and writing of this dissertation. I could not have imagined having a

better advisor and mentor for my Ph.D study. It was an eventful and inspiring time of my

life during my stay here at Athens. The kind of knowledge and experience I gained at

UGA in the field of Toxicology will have a great impact on my career. I am grateful to my

advisory committee members, Dr. Catherine A. White, Dr. Brian S. Cummings, Dr.

Michael G. Bartlett, and Dr. Randall Tackett for giving me suggestions and valuable

guidance at crucial junctures of my project. I thank them not only for their insightful

comments and encouragement, but also for the hard question which incented me to

widen my research from various perspectives. I would like to give special thanks to Dr.

Michael Bartlett and Dr. Brian Cummings for allowing me access to their lab and

equipment for sample preparation and analytical purposes. I sincerely thank Dr. Darren

Gullick for lending me immense support in analytical work. I cannot thank enough all my

colleagues in the lab, Dr. Jing Pang, Mr. Chen Chen, Mr. Tanzir Mortuza, and Mr.

Srinivasa Muralidhara for their stimulating discussions, and for all the fun we have had

in the last four years. They always created a friendly and positive ambience in the lab.

Finally, I am grateful to my parents, Ramesh Babu Amaraneni and Jyothi

Amaraneni, and my family who showered unconditional love and support. I specially

Page 8: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

vi

thank my wife, Sindhura Jonnalagadda, who stayed patient through all these years and

helped me focus on my program. Without my family, I could have achieved nothing.

Page 9: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

vii

TABLE OF CONTENTS

Page

ACKNOWLEDGEMENTS……………………………………………………………………...v

LIST OF TABLES……………………………………………………………………………....ix

LIST OF FIGURES……………………………………………………………………………...x

ABBREVIATIONS……………………………………………………………………………...xii

CHAPTER

1. INTRODUCTION AND LITERATURE REVIEW………………………………………..1

2. INFLUENCE OF MATURATION ON IN VIVO TISSUE TO PLASMA PARTITION

COEFFICIENTS OF DELTAMETHRIN, CIS- AND TRANS-PERMETHRIN IN

SPRAGUE-DAWLEY RATS……………………………………………………………..35

Abstract…………………………………………………………………………………36

Introduction……………………………………………………………………………..37

Materials and Methods………………………………………………………………..42

Results………………………………………………………………………………….47

Discussion……………………………………………………………………………...51

References……………………………………………………………………………..60

3. EFFECTS OF PLASMA PROTEIN BINDING AND MEMBRANE TRANSPORTERS

ON THE BLOOD BRAIN BARRIER PERMEABILITY OF DELTAMETHRIN………88

Abstract…………………………………………………………………………………89

Page 10: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

viii

Introduction……………………………………………………………………………..90

Materials and Methods………………………………………………………………..92

Results…………………………………………………………………………………95

Discussion……………………………………………………………………………..95

References……………………………………………………………………………101

4. BRAIN UPTAKE OF DELTAMETHRIN IN SPRAGUE DAWLEY RATS AS A

FUNCTION OF AGE DEPENDENT DIFFERENCES IN PLASMA PROTEIN

BINDING AND BBB PENETRATION…………………………………………………109

Abstract………………………………………………………………………………..110

Introduction……………………………………………………………………………112

Materials and Methods………………………………………………………………115

Results………………………………………………………………………………...121

Discussion…………………………………………………………………………….124

References……………………………………………………………………………134

5. SUMMARY………………………………………………………………………………161

APPENDIX……………………………………………………………………………………169

Page 11: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

ix

LIST OF TABLES

Page

Table 2.1: In Vitro Rat Plasma to Red Blood Cell (RBC) Partition Coefficients for

DLM (A), CIS (B) and TRANS (C)……..……………………………………………………69

Table 2.2: DLM (A), CIS (B) and TRANS (C) Mean Concentrations and Tissue:Plasma

Partition Coefficients for Adult Rat Tissues After 72-h Infusion…………………………71

Table 2.3: Comparison of DLM (A), CIS (B) and TRANS (C) Tissue:Plasma Partition

Coefficients with those Previously Published for Adult Rats.…………………………….72

Table 2.4: DLM (A), CIS (B) and TRANS (C) Mean Concentrations and Tissue:Plasma

Partition Coefficients for 21-day-old Rats After 48- and 72-h Infusions…………………75

Table 2.5: DLM (A), CIS (B) and TRANS (C) Mean Concentrations and Tissue:Plasma

Partition Coefficients for 15-day-old Rats After 48- and 72-h Infusions…………………78

Table 2.6: Comparison of DLM (A), CIS (B) and TRANS (C) Steady-State

Tissue:Plasma Partition Coefficients for Adults, 21-day, and 15-day-old Rats with those

Estimated from Single Dose Toxicokinetic Studies………………………………………81

Table 4.1: Relative Increase in the Brain Uptake of Different Concentrations of DLM in

PND 15 vs PND 21 vs Adults……………………………………………………………….158

Page 12: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

x

LIST OF FIGURES

Page

Figure 1.1: Structure of Deltamethrin and Permethrin…………………………………….31

Figure 1.2: Effect of Pyrethroids on Voltage Sensitive Sodium Channels in the CNS...32

Figure 1.3: General Metabolic Pathway of Pyrethroid Insecticides……………………...33

Figure 1.4: Different Means of Transport Across the BBB……………………………….34

Figure 2.1: Experimental Approach for In vivo Tissue:Plasma PC Experiment………..84

Figure 2.2: Comparison of Steady-State Tissue:Plasma PCs for DLM between PND 15,

PND 21 and Adult Rats after 72-h Infusion…………………………………………………85

Figure 2.3: Comparison of Steady-State Tissue:Plasma PCs for CIS between PND 15,

PND 21 and Adult Rats after 72-h Infusion…………………………………………………86

Figure 2.4: Comparison of Steady-State Tissue:Plasma PCs for TRANS between PND

15, PND 21 and Adult Rats after 72-h Infusion…………………………………………….87

Figure 3.1: Effect of HSA and CSA on the In vivo Brain Uptake of DLM……………...107

Figure 3.2: Effect of Mannitol on the In vivo Brain Association of DLM With and Without

4% HSA……………………………………………………………………………………….108

Figure 4.1: Effect of Time of Incubation on In vitro Binding of DLM to Human

Plasma………………………………………………………………………………………...144

Figure 4.2: Effect of Time of Incubation on In vitro Binding of DLM to HSA…………..145

Figure 4.3: Effect of Time of Incubation on In vitro Binding of DLM to LDL…………...146

Page 13: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

xi

Figure 4.4: Effect of Time of Incubation on In vitro Binding of DLM to HDL…………..147

Figure 4.5: Effect of Time of Incubation on In vitro Binding of DLM to Mixture of LDL

and HDL……………………………………………………………………………………….148

Figure 4.6: Effect of Binding to Physiological HSA Concentrations on the In vivo Brain

Uptake of DLM……………………………………………………………………………….149

Figure 4.7: In vitro Binding of 1 µM 14C-DLM to Different Concentrations of HSA…..150

Figure 4.8: Effect of Binding to Physiological LDL & HDL Concentrations on the In vivo

Brain Uptake of DLM…………………………………………………………………………151

Figure 4.9: In vitro Binding of 1 µm 14

C-DLM to Physiological Concentrations of LDL,

HDL, and their Mixture……………………………………………………………………….152

Figure 4.10: Effect of Binding to Lower, Normal and Higher Physiological LDL

Concentrations on the In vivo Brain Uptake of DLM……………………………………..153

Figure 4.11: In vitro Binding of 1 µM 14

C-DLM to Different Concentrations of LDL…..154

Figure 4.12: Effect of Binding to Rat Plasma of Different Age Groups on In vivo Brain

Uptake of DLM………………………………………………………………………………..155

Figure 4.13: Effect of Binding to Human Plasma of Different Age Groups on In vivo

Brain Uptake of DLM…………………………………………………………………………156

Figure 4.14: Effect of Maturity of BBB on In vivo Brain Uptake of DLM……………….157

Figure 4.15: Concentration Dependent Uptake of DLM in PND 15, PND 21, and Adult

SD Rats………………………………………………………………………………………..159

Figure 4.16: Effect of Flow Rate on In vivo Brain Uptake of DLM……………………...160

Page 14: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

xii

ABBREVIATIONS

3-PBA = 3-phenoxy benzoic acid

ABC = ATP-binding cassette

ANOVA = Analysis of Variance

AUC = Area under the Curve

BBB = Blood Brain Barrier

CEs = carboxylesterases

CIS = cis-permethrin

CNS = Central Nervous System

DLM = deltamethrin

fu = fraction unbound

hCEs = human carboxylesterases

hCMEC = human cerebral microvessel endothelial cells

HDL = high density lipoprotein

HSA = human serum albumin

Ko = infusion rate

LD = loading dose

LDL = low density lipoprotein

Log P/Log Ko/w = octanol to water partition coefficient

OAT = Organic Anion Transporters

Page 15: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

xiii

OATP = Organic Anion Transporting Polypeptide

OCT = Organic Cation Transporters

PBPK = physiologically based pharmacokinetic

PC = partition coefficient

PER = permethrin

P-gp = p-glycoprotein

PND = post-natal day

RBC = red blood cells

SC = subcutaneous

SD = Sprague-Dawley

TRANS = trans-permethrin

t1/2 = half-life

Page 16: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

1

CHAPTER 1

INTRODUCTION AND LITERATURE REVIEW

Background

Pyrethrum is a natural tan-colored syrupy extract with well-known insecticidal

properties produced from the flowers of Chrysanthemum cinerariaefolium (Seed, 1973;

Todd et al., 2003). Pyrethrins are the bioactive compounds of pyrethrum. The use of

pyrethrins in the United States dates back to the 1860’s (Casida, 1980; Schofield and

Crisafulli, 1980) even though they were first registered for use as insecticides in the

1950’s (Agency., 2006). Isobutenyl, furan, and allyl substituents of pyrethrins are highly

susceptible to photooxidation, making them not persistent enough for agricultural use

(Casida, 1980). Since pyrethrins break down easily when exposed to sunlight,

pyrethroids, which are synthetic analogs of pyrethrins were developed (Casida, 1980;

Soderlund et al., 2002). Combined with increased photostability and enhanced

insecticidal potency, pyrethroids are far more effective in their agricultural and

household usage compared to pyrethrins. Over the last decade, the use of pyrethroids

has dramatically increased in the U.S., Canada, and the European Union, due to the

U.S. EPA’s restrictions on use of organophosphates and due to their low environmental

persistence and relatively lower mammalian toxicity (Power and Sudakin, 2007;

Williams et al., 2008). By the mid-1990’s, 23% of the worldwide insecticide market was

pyrethroids (Casida and Quistad, 1998). Pyrethroids are often used in agricultural,

Page 17: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

2

veterinary, medical and household settings (Soderlund et al., 2002). They are applied to

a variety of crops for pest control and are also used to control vectors thereby promoting

public health. In household settings, they are often used as knockdown agents to

control houseflies and are also used in food storage areas. Medically, they are used for

the topical treatment of scabies, mites and headlice infestations of pets and humans.

Deltamethrin (DLM) is used to control mosquitoes in some tropical countries (Bradberry

et al., 2005).

Human exposure to pyrethroids albeit at very low levels, can occur through a

variety of routes like eating foods containing residues of pyrethroids, inhalation, skin

contact and accidental ingestion of pyrethroid products (Baker et al., 2000; Schettgen

et al., 2002). Pregnant women, infants, and children are widely exposed to pyrethroids

(Schettgen et al., 2002; Whyatt et al., 2002). Pyrethroid metabolites were found in 75%

of urine samples from a large German population without occupational exposure to the

insecticide (Heudorf et al., 2004). Infants and children are more frequently exposed to

pyrethroids compared to adults, due to their higher hand to mouth activity resulting in

ingestion of dust or soil contaminated with pyrethroids.

Structure of Pyrethroids

Pyrethroids are obtained from the structural modifications of pyrethrins, which are

esters of cyclopropanecarboxylic acid and a cyclopentenolone alcohol (Soderlund et al.,

2002). Structural modifications were done either on the acid or alcohol moiety to

enhance the photostability without the loss of insecticidal activity. Such modifications

include the replacement of chlorines for methyl groups in the acid moiety and 3-

Page 18: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

3

phenoxybenzyl for cyclopentenolone ring in the alcohol moiety (Soderlund et al., 2002).

These modifications significantly increased the photostability without the loss of

insecticidal activity. Pyrethroids were classified as Type I and Type II pyrethroids based

upon their chemical structure and toxic signs. Type II pyrethroids differ from Type I

pyrethroids in the presence of cyano substituent on the 3-phenoxybenzyl alcohol moiety

(Figure 1.1). Type II pyrethroids have enhanced insecticidal potency due to the

presence of the α-cyano substituent. Allethrin, permethrin, tetramethrin, and bifenthrin

are examples of Type I pyrethroids. Deltamethrin, cypermethrin, and esfenvalerate are

examples of Type II pyrethroids (Todd et al., 2003). Sixteen pyrethroids are registered

for use in the U.S. in agricultural and consumer products (Bryant and Bite, 2003).

Pyrethroids are highly lipophilic compounds and thus have very low water solubility

Their log Ko/w values are in the range of 5.7 - 7.6 (Laskowski, 2002). Due to their high

octanol-water partition coefficients, pyrethroids partition into lipids and organic matter

and bind with high affinity to sediments. Pyrethroids are highly soluble in organic

solvents such as alcohol, chlorinated hydrocarbons, and kerosene (Todd et al., 2003).

Mechanism of Action

The principal site of action of pyrethroids is the voltage-sensitive sodium channels

in the central nervous system (Figure 1.2). Pyrethroids exert neurotoxicity in mammals

by delaying the closure of sodium channel gates, which results in repetitive discharge of

nerve signals and eventually paralysis (Eells et al., 1992; Soderlund and Bloomquist,

1989; Soderlund et al., 2002). Type I and II pyrethroids differ in the prolongation of

channel open times which contributes to the differences in the clinical manifestations

Page 19: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

4

after acute exposure (Ray, 2001). The duration of modification of sodium currents by

type I pyrethroids lasts for a few milliseconds resulting in repetitive nerve discharges,

whereas that of type II pyrethroids, lasts for several seconds resulting in stimulus-

dependent nerve depolarization and blockage (Davies et al., 2007; Shafer et al., 2005;

Vijverberg and Bercken, 1982).

Acute exposure of laboratory animals to high doses of pyrethroids results in two

distinct toxic syndromes. Type I pyrethroids produce aggressive sparring,

hyperexcitation, fine tremors progressing to whole-body tremor, and paresthesia,

typically named as “T-syndrome”. Alternatively, Type II pyrethroids produce toxic signs

such as excessive salivation, pawning and burrowing, coarse tremors progressing to

choreoathetosis, and clonic seizures, collectively referred to as “CS-syndrome”

(Lawrence and Casida, 1982; Ray et al., 2000; Shafer et al., 2005). In general,

mammals are less susceptible to acute toxicity of pyrethroids due to their large body

size, poor dermal absorption, and rapid detoxification mechanisms (Bradberry et al.,

2005; Walters et al., 2009). The acute oral LD50 for deltamethrin given in corn oil was

reported to be 87 mg/kg in male rats (Myer, 1989), whereas for permethrin (45/55

cis/trans), it was reported to be 584 mg/kg (Metker et al., 1978). The average daily

intake of permethrin by a 70 kilogram adult male is estimated to be about 3.2

micrograms per day (Todd et al., 2003).

Metabolism of Pyrethroids

Pyrethroids are metabolized primarily via two pathways: hydrolysis of the central

ester bond by carboxylesterases; and oxidation at one or more sites in the acid or

Page 20: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

5

alcohol moieties by cytochrome P450-dependent monooxygenases (Soderlund et al.,

2002). These initial hydrolytic and oxidative products undergo conjugation with

glucuronides, sulfates, or amino acids prior to excretion (Figure 1.3). CYP 2C6 and

2C11 in rats and CYP 2C9 and 3A4 in humans are the major CYP 450 isoforms that are

involved in the oxidation of several pyrethroids (Godin et al., 2007; Scollon et al., 2009).

Similarly, human carboxylesterase 1 (hCE1) and hydrolase A are the predominant

carboxylesterases (CEs) that are involved in the hydrolysis of pyrethroids in humans

and rats, respectively. Significant quantities of CEs are present in the serum of rats,

whereas human serum is reported to have no esterase activity (Li et al., 2005).

Therefore, blood may be an important tissue for hydrolysis of pyrethroids in rats, but not

in humans (Godin et al., 2007). Immaturity of these metabolic enzymes is believed to

contribute significantly to increased systemic exposure and neurotoxic effects of

pyrethroids in younger age groups. Post-natal day 10 (PND 10) rats were more

susceptible to neurotoxicity of DLM than PND 40 rats, due to limited metabolism in PND

10 rats (Anand et al., 2006). DLM and cis-permethrin (CIS) are primarily metabolized by

oxidative pathways, whereas trans-permethrin (TRANS) is primarily metabolized by

esterase mediated hydrolysis (Anand et al., 2006; Scollon et al., 2009). Usually, the

trans-isomer of pyrethroids is more rapidly hydrolyzed than the cis-isomer (Yang et al.,

2009). The presence of the cyano group on the alcohol moiety of type II pyrethroids

makes these pyrethroids less prone to ester hydrolysis and P450 oxidation (Anand et

al., 2006).

Page 21: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

6

Tissue:plasma Partition Coefficients

Once a chemical is introduced into the body, the extent of partitioning into different

tissues determines its fate and plays an important role in assessing its toxicological

potential. The tissue:plasma partition coefficient (PC) is an important chemical-specific

input parameter for physiologically based pharmacokinetic (PBPK) models which is

used to predict the uptake and distribution kinetics of a chemical (Peyret et al., 2010).

Accurate tissue:plasma PCs help describe the distribution of a test compound in key

tissues of the body. A PC can be defined as the ratio of the concentration of a chemical

in two phases (i.e., tissue and plasma), once it reaches equilibrium. The systemic

deposition of lipid-soluble chemicals is largely dependent upon their solubility in different

organs (Poulin and Krishnan, 1995). It follows that these compounds will partition most

readily into tissues with the highest lipid content (Parham et al., 1997). Thus, a

compound’s octanol:water PC is an important property used in computational

approaches to estimate tissue:plasma PCs (Benfenati et al., 2003; Basak et al., 2004;

DeJongh et al., 1997; Poulin and Krishnan, 1995). Krishnan and his colleagues

observed that tissue deposition of lipid soluble compounds is complex and subsequently

took binding to blood proteins into account in their calculations to obtain more reliable

results. This strategy, coupled with data on tissues’ neutral and phospholipid content,

was utilized by Parham et al. to estimate tissue:blood PCs for all 209 polychlorinated

biphenyl (PCB) congeners (Parham et al., 1997). An algorithm was recently developed,

which took into account deposition in neutral lipids and binding to plasma proteins,

lipoproteins and hemoglobin, but not tissue proteins (Peyret et al., 2010).

Page 22: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

7

Tissue:plasma PCs may also be derived by direct measurement in vitro. PCs for

the PBPK model for triadimefon (log Ko/w=3.2) were obtained by measuring levels of the

conazole fungicide in saline and the tissue fraction of a spiked homogenate (Crowell et

al., 2011). Tissue:blood PCs were calculated by dividing the tissue:saline PCs by the

blood:saline PC. A similar approach was employed involving ultrafiltration for pesticides.

Pesticides with a log Kow≥1.7 could not be analyzed, as they were retained by the

filtration units tested. A solid phase micro extraction (SPME) method was utilized for

isolation of more lipophilic pesticides, but tissue:plasma PCs could not be determined

for those with a log Kow≥4, due to excessive partitioning into the SPME fiber (Tremblay

et al., 2012). The log Kow values for DLM and permethrin are 6.1 and 6.5, respectively

(Todd et al., 2003). Rat tissue:blood PCs have been measured for a series of very

lipophilic n-alkanes (Smith et al., 2005). The log Kow of dodecane (n-C12), the longest-

chain hydrocarbon measured was 6.1. The vial equilibration technique used, however,

is dependent upon volatilization of the analyte. Pyrethroids’ volatility is insufficient for

such vapor analyses.

Tissue:plasma PCs have been determined for lipid-soluble chemicals in several

instances by direct measurement in vivo. It is common for PBPK modelers to utilize

organ and plasma concentration data reported by other researchers to calculate PCs.

This practice was used for construction of models for PCBs (Lutz et al., 1977), lindane

(DeJongh and Blaauboer, 1997), chlordecone (Belfiore et al., 2007), and

polychlorinated dibenzo-p-dioxins and –furans (PCDD/Fs) (Maruyama et al., 2002).

Tissue:plasma PCs for pyrethroid PBPK models developed to date have not

relied on steady-state measurements. A PBPK model for DLM developed by

Page 23: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

8

Mirfazaelian et al. utilized time-course data from oral studies in rats. Tissue:plasma PCs

were calculated as the ratio of the area under the tissue DLM concentration versus time

curve to the plasma area under the curve (AUC) (Mirfazaelian et al., 2006). Another

PBPK model for DLM developed by Godin et al. (Godin et al., 2010) used the

computational procedure of Poulin and Theil (Poulin and Theil, 2000) to calculate PCs.

This procedure was based on tissue composition and described DLM’s expected

solubility in each tissue compartment. Similar to the approach of Mirfazaelian et al.

approach, the only PBPK model for permethrin developed to date has utilized time-

course data from oral studies in rats to estimate tissue:plasma PCs (Tornero-Velez et

al., 2012). However, they adopted published PCs for deltamethrin (DLM), adapting

some to fit their experimental kinetic data for CIS.

Age-Related Differences in Disposition

It is generally believed that infants and children are more susceptible to the

toxic effects of pesticides than adults. Newborns undergo significant growth and

maturational changes in physiological and biochemical processes. These changes have

the potential to significantly affect the disposition of chemicals and thus their tissue

dosimetry. The younger and more immature the subject is, the more different their

pharmacokinetic processes are from that of an adult (Bruckner, 2000). Developmental

changes in the gastro-intestinal (GI) tract such as changes in secretions, motility,

metabolism and transport can influence the rate and extent of absorption of a chemical

and thus its bioavailability (Strolin Benedetti et al., 2005). Maturational changes in

membrane permeability, plasma protein binding, cardiac output, and body content of

water and lipids may affect the distribution of a chemical in developing infants (Alcorn

Page 24: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

9

and McNamara, 2003). In general, immature neonates exhibit high membrane

permeability compared to adults. Plasma protein binding is lower in newborns and

young infants (Renwick, 1998). Factors such as lower plasma protein concentrations,

lower binding affinity and availability of competing endogenous substrates will result in

decreased binding in neonates. This results in an increased free fraction of the

chemical, thereby increasing its distribution into tissues. Newborns have higher total

body water (80-90%) and lower fat content (10-15%) compared to older children and

adults (Barrett et al., 2012; Lentner, 1981). Thus, volume of distribution of fat-soluble

chemicals will be relatively lower in newborns than in adults. In newborns, many

metabolic enzyme systems are immature and inefficient resulting in decreased

clearance of the chemical. This results in a higher plasma concentration of the

chemical. Even the renal clearance capacity of newborns is much lower when

compared to adults. Because of this complex pattern of maturational changes in the

pharmacokinetic processes, it is often difficult to predict the net effect of these changes

on the disposition of the chemical (Bruckner, 2000). Increased unbound fraction,

combined with the immature blood brain barrier (BBB) and higher cerebral blood flow

leads to greater partitioning of potential neurotoxins into the brain, thereby increasing

the susceptibility of neonates to neurotoxicity.

Age-related differences in the sensitivity of immature and adult rats to the

neurotoxicity of organophosphates have been clearly established. With regard to

malathion, Lu et al. reported that newborns exhibit the highest sensitivity to malathion

poisoning when compared to adults (Lu et al., 1965). It was also reported that lower

levels of hepatic CEs and P450-mediated dearylation play a major role in the increased

Page 25: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

10

sensitivity of juveniles to phosphorothionate toxicity (Atterberry et al., 1997). A similar

observation has been made pertaining to high doses of DLM. PND 11 and 21 pups

were found to be 16- and 7-times, respectively, more sensitive to acute neurotoxicity of

DLM than adults (Sheets et al., 1994). Sheets et al. reported that this increased

susceptibility of immature rats was most likely the result of lower capacity of metabolic

enzymes, which becomes quite apparent at acutely toxic doses. Thus, dispositional

factors rather than functional factors appear to contribute significantly to the relative

sensitivity of younger age groups to pyrethroid neurotoxicity.

Relying on animal studies for developing risk assessment of pyrethroids in

humans presents a problem. Not only are there significant differences in the enzymes

involved in the metabolism of pyrethroids, but also the rate at which these enzymes

mature is significantly different from humans. PBPK modeling is an important tool used

in risk analysis to predict early life sensitivity to pyrethroids. This tool is particularly

important when extrapolating across species and different age-groups. A PBPK model

is a unique framework which incorporates various age-specific and species-specific

pharmacokinetic factors that can impact the tissue dosimetry (Clewell et al., 2004). By

incorporating the age-specific differences, it allows us to modify a validated PBPK

model with adult pharmacokinetic data and use it for predicting concentration-time

courses in young humans. Thus, this modeling approach can be used successfully to

predict the relationship in humans between internal doses in the adult and internal

doses during early life using chemical-specific parameters. In order to validate the

accuracy of such predictions, proof-of-concept studies with representative pyrethroids

should be conducted in rats. The parameters in a PBPK model can be categorized into

Page 26: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

11

four types: exposure, physiological, partitioning, and metabolism (Clewell et al., 2007). It

is important to obtain accurate measurements of physiological (tissue and fat volumes,

circulation), metabolism and partition coefficient parameters to develop a reliable PBPK

model.

Objective and Significance

Tissue:plasma PCs are key input parameters for PBPK models. Validated PBPK

models can be used for reliable predictions of pyrethroid internal dosimetry in humans.

Limited data are available in the literature from which to determine PCs for pyrethroids.

An important implication of that data is that it is rather estimated and not accurately

measured at steady-state. No studies till date have measured the tissue:plasma PCs for

pyrethroids at steady-state or assessed differences in the distribution of these

compounds between immature and adult rats.

Therefore, the overall objective of this project is to gain an insight into the systemic

distribution of representative pyrethroid insecticides and to understand differences

between the younger age-groups and adults in the pesticides disposition. The

knowledge gained from these studies can be useful in risk assessment of pyrethroids in

human sub-populations. Also the other scientists can use this information in the future

to predict the distribution of other pesticides with similar physicochemical properties in

different age-groups.

Blood-Brain-Barrier

The BBB is a selective ‘diffusion’ barrier, which restricts the entry of most

compounds from blood to brain. The BBB plays an important role in maintaining the

Page 27: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

12

homeostasis of the brain by regulating ionic and fluid movements between blood and

brain. It also helps supply the brain with essential nutrients and protects the brain from

circulating toxins and waste products. Endothelial cells, astrocyte end feet and pericytes

constitute the cellular elements of the brain microvasculature that compose the BBB

(Ballabh et al., 2004). The endothelial cells at the BBB are characterized by a

continuous basement membrane, extensive tight junctions, absence of fenestrations

and sparse pinocytotic vesicular transport (Sage, 1982). These characteristics

differentiate the endothelia of cerebral capillaries from that of non-neural tissues.

Because of the continuous tight junctions, the endothelium at the BBB behave like a

plasma membrane (Sage, 1982). The tight junctions at the BBB are 50-100 times tighter

than peripheral microvessels and prevent the entry of small hydrophilic molecules by

limiting paracellular transport (Lawther et al., 2011). Therefore, the transport across the

BBB is effectively limited to selective transcellular mechanisms. It is generally believed

that the compounds with low molecular weight, high lipid solubility, low plasma protein

binding, and a low degree of ionization at physiological pH diffuse freely across the BBB

along their concentration gradient and attain rapid equilibrium. On the other hand,

compounds with low lipid solubility, high polar surface area, high plasma protein

binding, high molecular weight and a high degree of ionization at physiological pH have

restricted and slow BBB permeability (Clark, 2003) .

The presence of specific influx and efflux transporters on the apical and

basolateral membranes of the endothelia further regulates the bidirectional transcellular

transport across the BBB, thus providing a selective ‘transport’ barrier (Figure 1.4)

(Abbott et al., 2006). Carrier mediated influx via facilitative diffusion is a passive process

Page 28: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

13

that can transport many essential polar molecules such as glucose, amino acids and

nucleosides into the central nervous system (CNS). Facilitative diffusion is an energy

independent process, where the solute molecules bind to specific solute carriers (SLCs)

and then move along their concentration gradients. On the other hand, efflux

transporters (ABC transporters) are primarily active and energy-dependent transporters

that intercept some of the passively diffusing solutes and pump them out of the

endothelial cells (Abbott et al., 2010). It is the orientation and interplay of these

transporters at the BBB that determines the direction of the transport (blood to brain or

brain to blood) and the extent to which a chemical gains access to the CNS (Lee et al.,

2001; Löscher and Potschka, 2005; Sun et al., 2003). Endocytosis is another important

transport mechanism by which large molecules such as proteins and peptides enter the

CNS. Endocytosis can be either receptor mediated (RME) or adsorptive mediated

(AME) and contributes to selective uptake of macromolecules (Abbott et al., 2010).

RME transports substances such as insulin, transferrin, and insulin like growth factor

into the CNS and is highly energy dependent process (Lawther et al., 2011).

Ontogeny of Blood-Brain-Barrier

The BBB in newborns is immature and is poorly formed and leaky. The immature

BBB in newborns contributes to higher brain levels of chemicals (Arya et al., 2006).

Arya et al. reported that the brain uptake of glucocorticoids in neonatal rats is

significantly higher than adults due to an immature BBB. The development of BBB is a

gradual process in humans and it reaches adult capabilities by approximately 6 months

of age (Watson et al., 2006). The fully differentiated (i.e., mature) BBB consisted of

highly specialized endothelial cells, large number of pericytes imbedded in the

Page 29: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

14

basement membrane, perivascular macrophages and overlapping astrocytic end feet

(Engelhardt, 2003). Behnsen (1905) found that upon injection of very high

concentrations of trypan blue, mice of 4 weeks of age incorporated more dye compared

to mice of 5-8 weeks of age. This suggests that the BBB is more permeable in younger

mice compared to adults. Clark et al. reported that the BBB permeability is high at birth

and decreases in the first few weeks after birth (Clark et al., 1993). The BBB was not

fully developed structurally and was not fully functional until about the time of weaning

(Schulze and Firth, 1992). A clearly delineated basement membrane was absent in

embryonic capillaries and was not fully developed until PND 16. Schulze and Firth

suggested that maturation of interendothelial clefts is accompanied by establishment of

a characteristic ratio of ‘narrow zone’ (complex tight junctions) to ‘wide zone’ and

disappearance of membrane separations larger than 20 nm. While tight junctional

contacts in immature BBB were described as having ‘touching’ outer leaflets, adult BBB

appear to have ‘fused’ outer leaflets. Leaky interendothelial junctions in the developing

brain results in high permeability. Apart from gradual decrease in unfused endothelial

cell outer leaflets, junctional clefts also decrease and expanded junctional clefts

(paracellular channels) virtually disappear with age in parallel to decreased permeability

(Stewart and Hayakawa, 1987). Thus, developmental tightening of the BBB is primarily

characterized by structural changes at tight junctional contacts. Like most other

structures and physiological functions, the BBB is more mature at birth in humans than

in rodents. Its rate of maturation in human infants is maximal during the third month of

life. The rate of change progressively diminishes thereafter, until the fully developed

BBB is achieved by 1 year of age (Statz and Felgenhauer, 1983).

Page 30: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

15

Apart from the morphological changes, specific transporters at the BBB also

undergo changes in their expression with age. Thus, ontogeny of transporters at the

luminal and abluminal side of the membrane can affect the amount of chemical that

enters the CNS. P-gp was first detected on PND 7, reaches 25% of the adult by PND 10

and then reaches plateau by PND 28 (Matsuoka et al., 1999). BCRP mRNA levels were

high at birth and decreased to adult levels by PND 42. For OCT1, OCT2 and OAT1,

mRNA levels peaked around PND 1 to 4 with limited expression before birth and

throughout development. For some other transporters such as multi-drug resistance 1a

(MDR1a), MRP3, OAT2 and OAT3, mRNA expression remained more or less constant

throughout the development (de Zwart et al., 2008).

Since brain is the target organ of potential toxicity for pyrethroids, it is of great

importance not only to accurately measure the permeability of pyrethroids, but also to

understand various factors that contribute to their penetration into the brain. Even

though DLM is a highly lipophilic compound (log Ko/w = 6.1), its entry into the brain is

restricted by its high molecular weight (505.2) and high plasma protein binding (80%).

Plasma protein binding plays a very important role in drug transport since only unbound

drug is able to penetrate the membrane. Therefore, the free fraction of drug in the

capillary blood is an important determinant of drug entry into the brain. There is a lack of

understanding of the effect of plasma protein binding on brain uptake of pyrethroids and

the absence of an accurate quantitative model that appropriately describes this effect.

The plasma protein binding of many drugs is believed to be age dependent as

mentioned in the previous section (1.6). Total plasma protein concentrations increased

from 2.5 g/dL at birth to about 5.2 g/dL in adult rats and the corresponding albumin

Page 31: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

16

concentration increased from 1.8 g/dL to 3.8 g/dL (de Zwart et al., 2008). Thus, the

difference in plasma protein concentrations and the binding between newborns and

adults can alter the brain uptake of pyrethroids. Thus, it is also of great significance to

evaluate the effect of age-dependent changes in plasma protein binding and BBB

maturity on the permeability of pyrethroids. Therefore, as a part of the overall project,

BBB permeability of DLM will be determined as a function of age-dependent changes in

BBB integrity and plasma protein binding.

Hypothesis of Dissertation

It is hypothesized that the distribution and brain uptake of pyrethroids in immature rats

will be significantly different from that in adult rats due to differences in plasma protein

binding and BBB permeability.

Overview of Experimental Plan of Dissertation

1. To determine in vivo tissue:plasma partition coefficients of DLM, CIS and TRANS

in PND 15, PND 21 and adult rats.

2. To selectively investigate the role of a) plasma protein binding; b) membrane

transporters; c) BBB – in limiting the uptake of pyrethroids into the brain.

3. To evaluate the brain uptake of DLM as a function of age-dependent changes in

BBB integrity and plasma protein binding.

Page 32: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

17

REFERENCES

Abbott, N. J., Patabendige, A. A., Dolman, D. E., Yusof, S. R., and Begley, D. J. (2010).

Structure and function of the blood–brain barrier. Neurobiology of disease 37(1), 13-25.

Abbott, N. J., Rönnbäck, L., and Hansson, E. (2006). Astrocyte–endothelial interactions

at the blood–brain barrier. Nature Reviews Neuroscience 7(1), 41-53.

Agency., U. E. P. (2006). Reregistration eligibility decision for Pyrethrins. Case no.

2580. In (doi. US Environmental Protection Agency Washington, DC.

Alcorn, J., and McNamara, P. J. (2003). Pharmacokinetics in the newborn. Advanced

drug delivery reviews 55(5), 667-686.

Anand, S. S., Kim, K.-B., Padilla, S., Muralidhara, S., Kim, H. J., Fisher, J. W., and

Bruckner, J. V. (2006). Ontogeny of hepatic and plasma metabolism of deltamethrin in

vitro: role in age-dependent acute neurotoxicity. Drug metabolism and disposition 34(3),

389-397.

Arya, V., Demarco, V. G., Issar, M., and Hochhaus, G. (2006). Contrary to adult,

neonatal rats show pronounced brain uptake of corticosteroids. Drug metabolism and

disposition 34(6), 939-942.

Page 33: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

18

Atterberry, T. T., Burnett, W. T., and Chambers, J. E. (1997). Age-related differences in

parathion and chlorpyrifos toxicity in male rats: target and nontarget esterase sensitivity

and cytochrome P450-mediated metabolism. Toxicology and applied pharmacology

147(2), 411-418.

Baker, S. E., Barr, D. B., Driskell, W. J., D BEESON, M., and Needham, L. L. (2000).

Quantification of selected pesticide metabolites in human urine using isotope dilution

high-performance liquid chromatography/tandem mass spectrometry. Journal of

Exposure Science and Environmental Epidemiology 10, 789-798.

Ballabh, P., Braun, A., and Nedergaard, M. (2004). The blood–brain barrier: an

overview: structure, regulation, and clinical implications. Neurobiology of disease 16(1),

1-13.

Barrett, J., Alberighi, O. D. C., Läer, S., and Meibohm, B. (2012). Physiologically based

pharmacokinetic (PBPK) modeling in children. Clinical Pharmacology & Therapeutics

92(1), 40-49.

Belfiore, C. J., Yang, R. S., Chubb, L. S., Lohitnavy, M., Lohitnavy, O. S., and

Andersen, M. E. (2007). Hepatic sequestration of chlordecone and hexafluoroacetone

evaluated by pharmacokinetic modeling. Toxicology 234(1), 59-72.

Page 34: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

19

Benfenati, E., Gini, G., Piclin, N., Roncaglioni, A., and Varı, M. (2003). Predicting log< i>

P</i> of pesticides using different software. Chemosphere 53(9), 1155-1164.

Bradberry, S. M., Cage, S. A., Proudfoot, A. T., and Vale, J. A. (2005). Poisoning due to

pyrethroids. Toxicological reviews 24(2), 93-106.

Bruckner, J. V. (2000). Differences in sensitivity of children and adults to chemical

toxicity: the NAS panel report. Regulatory toxicology and pharmacology 31(3), 280-285.

Bryant, R., and Bite, M. (2003). Global insecticide directory. In (doi. Orpington, Kent,

UK: Agranova.

C Basak, S., Mills, D., El-Masri, H. A., Mumtaz, M. M., and Hawkins, D. M. (2004).

Predicting blood: air partition coefficients using theoretical molecular descriptors.

Environmental toxicology and pharmacology 16(1), 45-55.

Casida, J. E. (1980). Pyrethrum flowers and pyrethroid insecticides. Environmental

health perspectives 34, 189.

Casida, J. E., and Quistad, G. B. (1998). Golden age of insecticide research: past,

present, or future? Annual review of entomology 43(1), 1-16.

Clark, D. E. (2003). In silico prediction of blood–brain barrier permeation. Drug

discovery today 8(20), 927-933.

Page 35: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

20

Clark, J., Bates, T., Cullingford, T., and Land, J. (1993). Development of enzymes of

energy metabolism in the neonatal mammalian brain. Developmental neuroscience

15(3-5), 174-180.

Clewell, H. J., Gentry, P. R., Covington, T. R., Sarangapani, R., and Teeguarden, J. G.

(2004). Evaluation of the potential impact of age-and gender-specific pharmacokinetic

differences on tissue dosimetry. Toxicological Sciences 79(2), 381-393.

Clewell, H. J., Reddy, M. B., Lave, T., and Andersen, M. E. (2007). Physiologically

based pharmacokinetic modeling. Pharmaceutical Sciences Encyclopedia doi.

Crowell, S. R., Henderson, W. M., Kenneke, J. F., and Fisher, J. W. (2011).

Development and application of a physiologically based pharmacokinetic model for

triadimefon and its metabolite triadimenol in rats and humans. Toxicology letters 205(2),

154-162.

Davies, T., Field, L., Usherwood, P., and Williamson, M. (2007). DDT, pyrethrins,

pyrethroids and insect sodium channels. IUBMB life 59(3), 151-162.

de Zwart, L., Scholten, M., Monbaliu, J. G., Annaert, P. P., Van Houdt, J. M., Van den

Wyngaert, I., De Schaepdrijver, L. M., Bailey, G. P., Coogan, T. P., and Coussement,

Page 36: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

21

W. C. (2008). The ontogeny of drug metabolizing enzymes and transporters in the rat.

Reproductive Toxicology 26(3), 220-230.

DeJongh, J., and Blaauboer, B. J. (1997). Simulation of lindane kinetics in rats.

Toxicology 122(1), 1-9.

DeJongh, J., Verhaar, H. J., and Hermens, J. L. (1997). A quantitative property-property

relationship (QPPR) approach to estimate in vitro tissue-blood partition coefficients of

organic chemicals in rats and humans. Archives of toxicology 72(1), 17-25.

Eells, J. T., Bandettini, P. A., Holman, P. A., and Propp, J. M. (1992). Pyrethroid

insecticide-induced alterations in mammalian synaptic membrane potential. Journal of

Pharmacology and Experimental Therapeutics 262(3), 1173-1181.

Engelhardt, B. (2003). Development of the blood-brain barrier. Cell and tissue research

314(1), 119-129.

Godin, S. J., Crow, J. A., Scollon, E. J., Hughes, M. F., DeVito, M. J., and Ross, M. K.

(2007). Identification of rat and human cytochrome p450 isoforms and a rat serum

esterase that metabolize the pyrethroid insecticides deltamethrin and esfenvalerate.

Drug Metabolism and Disposition 35(9), 1664-1671.

Page 37: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

22

Godin, S. J., DeVito, M. J., Hughes, M. F., Ross, D. G., Scollon, E. J., Starr, J. M.,

Setzer, R. W., Conolly, R. B., and Tornero-Velez, R. (2010). Physiologically based

pharmacokinetic modeling of deltamethrin: development of a rat and human diffusion-

limited model. Toxicological Sciences doi, kfq051.

Heudorf, U., Angerer, J., and Drexler, H. (2004). Current internal exposure to pesticides

in children and adolescents in Germany: urinary levels of metabolites of pyrethroid and

organophosphorus insecticides. International archives of occupational and

environmental health 77(1), 67-72.

Laskowski, D. A. (2002). Physical and chemical properties of pyrethroids. In Reviews of

environmental contamination and toxicology (doi, pp. 49-170. Springer.

Lawrence, L. J., and Casida, J. E. (1982). Pyrethroid toxicology: mouse intracerebral

structure-toxicity relationships. Pesticide Biochemistry and Physiology 18(1), 9-14.

Lawther, B. K., Kumar, S., and Krovvidi, H. (2011). Blood–brain barrier. Continuing

Education in Anaesthesia, Critical Care & Pain 11(4), 128-132.

Lee, G., Dallas, S., Hong, M., and Bendayan, R. (2001). Drug transporters in the central

nervous system: brain barriers and brain parenchyma considerations. Pharmacological

reviews 53(4), 569-596.

Page 38: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

23

Lentner, C. (1981). Geigy Scientific tables. Vol. 1, Units of measurement, body fluids,

composition of the body, nutrition. Ciba-Geigy Basle.

Li, B., Sedlacek, M., Manoharan, I., Boopathy, R., Duysen, E. G., Masson, P., and

Lockridge, O. (2005). Butyrylcholinesterase, paraoxonase, and albumin esterase, but

not carboxylesterase, are present in human plasma. Biochemical pharmacology 70(11),

1673-1684.

Löscher, W., and Potschka, H. (2005). Blood-brain barrier active efflux transporters:

ATP-binding cassette gene family. NeuroRx 2(1), 86-98.

Lu, F., Jessup, D., and Lavallee, A. (1965). Toxicity of pesticides in young versus adult

rats. Food and cosmetics toxicology 3, 591-596.

Lutz, R., Dedrick, R., Matthews, H., Eling, T., and Anderson, M. (1977). A preliminary

pharmacokinetic model for several chlorinated biphenyls in the rat. Drug metabolism

and disposition 5(4), 386-396.

Maruyama, W., Yoshida, K., Tanaka, T., and Nakanishi, J. (2002). Determination of

tissue–blood partition coefficients for a physiological model for humans, and estimation

of dioxin concentration in tissues. Chemosphere 46(7), 975-985.

Page 39: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

24

Matsuoka, Y., Okazaki, M., Kitamura, Y., and Taniguchi, T. (1999). Developmental

expression of P‐glycoprotein (multidrug resistance gene product) in the rat brain.

Journal of neurobiology 39(3), 383-392.

Metker, L., Angerhofer, R., Pope, C., and Swentzel, K. (1978). Toxicological evaluation

of 3-(phenoxyphenyl) methyl (±)-cis, trans-3-(2, 2-dichloroethenyl)-2, 2-

dimethylcyclopropane carboxylate (Permethrin).

Mirfazaelian, A., Kim, K.-B., Anand, S. S., Kim, H. J., Tornero-Velez, R., Bruckner, J. V.,

and Fisher, J. W. (2006). Development of a physiologically based pharmacokinetic

model for deltamethrin in the adult male Sprague-Dawley rat. Toxicological Sciences

93(2), 432-442.

Myer, J. (1989). Acute oral toxicity study of deltamethrin in rats. Hoechst-Roussel Agri-

Vet Company Study doi: (327-122).

Parham, F., Kohn, M., Matthews, H., DeRosa, C., and Portier, C. (1997). Using

structural information to create physiologically based pharmacokinetic models for all

polychlorinated biphenyls: I. Tissue: blood partition coefficients. Toxicology and applied

pharmacology 144(2), 340-347.

Page 40: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

25

Peyret, T., Poulin, P., and Krishnan, K. (2010). A unified algorithm for predicting

partition coefficients for PBPK modeling of drugs and environmental chemicals.

Toxicology and applied pharmacology 249(3), 197-207.

Poulin, P., and Krishnan, K. (1995). A biologically-based algorithm for predicting human

tissue: blood partition coefficients of organic chemicals. Human & experimental

toxicology 14(3), 273-280.

Poulin, P., and Theil, F. P. (2000). A priori prediction of tissue: plasma partition

coefficients of drugs to facilitate the use of physiologically‐based pharmacokinetic

models in drug discovery. Journal of pharmaceutical sciences 89(1), 16-35.

Power, L. E., and Sudakin, D. L. (2007). Pyrethrin and pyrethroid exposures in the

United States: a longitudinal analysis of incidents reported to poison centers. Journal of

medical toxicology 3(3), 94-99.

Ray, D. (2001). Pyrethroid insecticides: mechanisms of toxicity, systemic poisoning

syndromes, paresthesia, and therapy. Handbook of pesticide toxicology 2, 1289-1303.

Ray, D. E., Ray, D., and Forshaw, P. J. (2000). Pyrethroid insecticides: poisoning

syndromes, synergies, and therapy. Journal of Toxicology: Clinical Toxicology 38(2),

95-101.

Page 41: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

26

Renwick, A. (1998). Toxicokinetics in infants and children in relation to the ADI and TDI.

Food Additives & Contaminants 15(S1), 17-35.

Sage, M. (1982). Blood-brain barrier: phenomenon of increasing importance to the

imaging clinician. American Journal of Roentgenology 138(5), 887-898.

Schettgen, T., Heudorf, U., Drexler, H., and Angerer, J. (2002). Pyrethroid exposure of

the general population—is this due to diet. Toxicology letters 134(1), 141-145.

Schofield, E. K., and Crisafulli, S. (1980). A safer insecticide for herbarium use. Brittonia

32(1), 58-62.

Schulze, C., and Firth, J. A. (1992). Interendothelial junctions during blood-brain barrier

development in the rat: morphological changes at the level of individual tight junctional

contacts. Developmental brain research 69(1), 85-95.

Scollon, E. J., Starr, J. M., Godin, S. J., DeVito, M. J., and Hughes, M. F. (2009). In vitro

metabolism of pyrethroid pesticides by rat and human hepatic microsomes and

cytochrome p450 isoforms. Drug Metabolism and Disposition 37(1), 221-228.

Seed, P. (1973). Pyrethrum: the natural insecticide doi.

Page 42: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

27

Shafer, T. J., Meyer, D. A., and Crofton, K. M. (2005). Developmental neurotoxicity of

pyrethroid insecticides: critical review and future research needs. Environmental health

perspectives 113(2), 123.

Sheets, L. P., Doherty, J. D., Law, M. W., Reiter, L. W., and Crofton, K. M. (1994). Age-

dependent differences in the susceptibility of rats to deltamethrin. Toxicology and

applied pharmacology 126(1), 186-190.

Smith, A., Campbell, J., Keys, D., and Fisher, J. (2005). Rat tissue and blood partition

coefficients for n-alkanes (C8 to C12). International journal of toxicology 24(1), 35-41.

Soderlund, D. M., and Bloomquist, J. R. (1989). Neurotoxic actions of pyrethroid

insecticides. Annual review of entomology 34(1), 77-96.

Soderlund, D. M., Clark, J. M., Sheets, L. P., Mullin, L. S., Piccirillo, V. J., Sargent, D.,

Stevens, J. T., and Weiner, M. L. (2002). Mechanisms of pyrethroid neurotoxicity:

implications for cumulative risk assessment. Toxicology 171(1), 3-59.

Statz, A., and Felgenhauer, K. (1983). Development of the Blood‐CSF Barrier.

Developmental Medicine & Child Neurology 25(2), 152-161.

Page 43: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

28

Stewart, P., and Hayakawa, E. (1987). Interendothelial junctional changes underlie the

developmental ‘tightening’of the blood-brain barrier. Developmental Brain Research

32(2), 271-281.

Strolin Benedetti, M., Whomsley, R., and Baltes, E. L. (2005). Differences in absorption,

distribution, metabolism and excretion of xenobiotics between the paediatric and adult

populations doi.

Sun, H., Dai, H., Shaik, N., and Elmquist, W. F. (2003). Drug efflux transporters in the

CNS. Advanced drug delivery reviews 55(1), 83-105.

Todd, G. D., Wohlers, D., and Citra, M. J. (2003). Toxicological profile for pyrethrins and

pyrethroids. Agency for Toxic Substances and Disease Registry GA.

Tornero-Velez, R., Davis, J., Scollon, E., Starr, J. M., Setzer, R. W., Goldsmith, M.,

Chang, D., Xue, J., Zartarian, V., and DeVito, M. J. (2012). A pharmacokinetic model of

cis-and trans-permethrin disposition in rats and humans with aggregate exposure

application. Toxicological Sciences doi, kfs236.

Tremblay, R. T., Kim, D., and Fisher, J. W. (2012). Determination of tissue to blood

partition coefficients for nonvolatile herbicides, insecticides, and fungicides using

negligible depletion solid-phase microextraction (nd-SPME) and ultrafiltration. Journal of

Toxicology and Environmental Health, Part A 75(5), 288-298.

Page 44: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

29

VIJVERBERG, H. P., and BERCKEN, J. V. D. (1982). Action of pyrethroid insecticides

on the vertebrate nervous system. Neuropathology and applied neurobiology 8(6), 421-

440.

Walters, J. K., Boswell, L. E., Green, M. K., Heumann, M. A., Karam, L. E., Morrissey,

B. F., and Waltz, J. E. (2009). Pyrethrin and pyrethroid illnesses in the Pacific

Northwest: a five-year review. Public Health Reports 124(1), 149.

Watson, R. E., DeSesso, J. M., Hurtt, M. E., and Cappon, G. D. (2006). Postnatal

growth and morphological development of the brain: a species comparison. Birth

Defects Research Part B: Developmental and Reproductive Toxicology 77(5), 471-484.

Whyatt, R. M., Camann, D. E., Kinney, P. L., Reyes, A., Ramirez, J., Dietrich, J., Diaz,

D., Holmes, D., and Perera, F. P. (2002). Residential pesticide use during pregnancy

among a cohort of urban minority women. Environmental health perspectives 110(5),

507.

Williams, M. K., Rundle, A., Holmes, D., Reyes, M., Hoepner, L. A., Barr, D. B.,

Camann, D. E., Perera, F. P., and Whyatt, R. M. (2008). Changes in pest infestation

levels, self-reported pesticide use, and permethrin exposure during pregnancy after the

2000–2001 US Environmental Protection Agency restriction of organophosphates.

Environ Health Perspect 116(12), 1681-1688.

Page 45: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

30

Yang, D., Wang, X., Chen, Y.-t., Deng, R., and Yan, B. (2009). Pyrethroid insecticides:

isoform-dependent hydrolysis, induction of cytochrome P450 3A4 and evidence on the

involvement of the pregnane X receptor. Toxicology and applied pharmacology 237(1),

49-58.

Page 46: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

31

A Deltamethrin

B Permethrin

Figure 1.1. Structure of Deltamethrin (A) and Permethrin (B). Adapted from Soderlund

et al. (2002). Pyrethroids are esters of a cyclopropanecarboxylic acid and a

cyclopentenolone alcohol. Addition of α-cyano group at the benzylic carbon atom gives

type II pyrethroids such as deltamethrin.

Page 47: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

32

Figure 1.2. Effect of Pyrethroids on Voltage Sensitive Sodium Channels in the CNS.

Adapted from Shafer et al. (2005). Pyrethroids delay the inactivation of voltage sensitive

sodium channels allowing more sodium ions to cross and depolarize the neuronal

membrane.

Page 48: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

33

Figure 1.3. General Metabolic Pathway of Pyrethroid Insecticides. Adapted from Kim et

al. (2010). The initial metabolism of the parent compound results from the actions of

either esterases at the central ester bond or CYP 450 mediated oxidation at one or

more sites in the acid or alcohol moieties. The metabolites from hydrolytic or oxidative

actions are further conjugated with sulfates or glucuronides prior to excretion.

Page 49: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

34

Figure 1.4. Different Means of Transport across the BBB. Adapted from Abbott et al.

(2010). Small, lipid soluble, non-polar molecules passively diffuse across the BBB.

Presence of various efflux and influx transports on both the apical and basolateral

membranes of endothelial cells can be observed from the figure.

Page 50: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

35

CHAPTER 2

INFLUENCE OF MATURATION ON IN VIVO TISSUE TO PLASMA PARTITION

COEFFICIENTS OF DELTAMETHRIN, CIS- AND TRANS-PERMETHRIN IN

SPRAGUE-DAWLEY RATS1

1Manoj A., Brian S.C., Jing P., Srinivasa M., Tanzir B.M., Darren G., Catherine A.W., and James V.B. To be submitted to Journal of Pharmaceutical Sciences.

Page 51: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

36

ABSTRACT

The major objective of this investigation was to develop a reliable approach for

determining in vivo partition coefficients (PCs) for deltamethrin (DLM), cis-permethrin

(CIS) and trans-permethrin (TRANS) in adult and young Sprague-Dawley (SD) rats.

Adult, PND 21 and PND 15 rats were infused with a predetermined amount of DLM, CIS

or TRANS via a sc osmotic pump for 48 or 72-h. Adult and PND 21 rats also received

an oral loading dose. Systemic steady-state, or equilibrium was attained in each age-

group within 72-h of the protocol. DLM, CIS and TRANS were all distributed to tissues

according to their neutral lipid content, with adipose tissue exhibiting much higher

tissue:plasma PCs than skeletal muscle, liver or brain. Liver:plasma and brain:plasma

PCs were consistently lower than unity. Tissue:plasma PCs were generally higher for

CIS than for DLM & TRANS, especially in immature rats. CIS is metabolized through

much more slower oxidative processes, which may result in significantly higher

circulating levels. This appears to saturate plasma binding and thus, might have

resulted in greater tissue deposition of CIS. CIS tissue:plasma PCs were found to be

inversely related to the rats’ age. This was also true for TRANS, although TRANS

brain:plasma PCs were comparable in immature and mature animals. Interestingly,

although DLM tissue:plasma PCs in PND 15 rats were significantly higher as compared

to adults, PCs in PND 21 rats were consistently lower than that of adults. These results

suggest that age-dependent partitioning may play an important role in determining the

kinetics and ensuing risks of these commonly utilized insecticides.

Page 52: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

37

INTRODUCTION

Pyrethroids, synthetic derivatives of naturally occurring pyrethrins, are active

components of household pesticide sprays, insect repellents, pet shampoos and human

lice and scabies treatments. These chemicals are also applied to agricultural fields,

commercial nurseries, residential structures and lawns and gardens. Permethrin (PER),

marketed as a mixture of its isomers CIS and TRANS, is the most frequently utilized

insecticide in the U.S (Barr et al., 2010). Thus, it is not surprising that a large proportion

of the general population in the U.S. and the E.U. are frequently exposed to such

chemicals. Three-phenoxy benzoic acid (3-PBA), a metabolite common to a number of

pyrethroids, was detected in 70% of over 5,000 people monitored in the U.S. in the

National Health and Nutrition Examination Survey (NHANES) (Barr et al., 2010).

Children exhibited significantly higher 3-PBA levels than adolescents or adults. Morgan

et al. (Morgan, 2012) compiled data from 15 published studies of pyrethroid exposure of

children in day-care centers and homes. Seven different pyrethroids were found in

dusts, floor wipes, air and urine samples. PER was the most frequently detected.

Ingestion from dusts and pets by hand-to-mouth activity was the most common source

of exposure. Consumption of the very low levels in foods was of secondary importance

(Lu et al., 2006). Substantially high doses of pyrethroids can be acutely neurotoxic,

though the potency of different pyrethroids varies widely (Wolansky et al., 2006). The

parent compounds are the proximate neuroactive moieties. Their primary mechanism of

action on neurons is interference with voltage-gated sodium channels, resulting in

stimulus-dependent nerve depolarization and block (Soderlund, 2012). The very high

lipophilicity (i.e., Log Ps = 4.6 – 7.1) of pyrethroids significantly impacts their

Page 53: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

38

pharmacokinetics. The chemicals appear to be poorly absorbed from the GI tract,

despite their lipid solubility. Oral bioavailability of DLM in rats is reported to be just 14-

16% (Anadon et al., 1996; Kim et al., 2008). DLM, CIS and TRANS are highly bound to

rat and human plasma proteins and lipoproteins (Sethi et al., 2014). Systemic

distribution of DLM and PER in rats is apparently governed largely by tissues’ fat

content. Pyrethroids, as a chemical class, readily undergo metabolic inactivation via

oxidation in the liver by cytochrome (CYP) P450s and hydrolysis by plasma and/or liver

carboxylesterases (CEs) in rats and humans. Species- and/or age-specific metabolic

rate constants have been published for DLM, CIS and TRANS (Anand et al., 2006;

Godin et al., 2006; Tornero-Velez et al., 2012).

PCs are important determinants of the systemic deposition and ensuing biological

effects of xenobiotics. A tissue:plasma PC can be defined as the ratio of concentrations

of a chemical in two phases once equilibrium is attained. As partitioning into tissues

substantially influences the pharmacokinetics of xenobiotics, PCs are an essential input

parameter for physiologically based pharmacokinetic (PBPK) models. PBPK models are

finding increasing use in predicting the internal dosimetry (i.e., plasma and tissue

concentration time-courses) of drugs (Rowland et al., 2011; Shardlow et al., 2013) and

other chemicals (Andersen, 2003; Lipscomb et al., 2012). PBPK models are also being

developed to predict target organ dosimetry and associated health risks of commonly-

used insecticides, including organophosphates and pyrethroids. Several first-generation

models have been published for DLM and PER, two widely-utilized pyrethroids. In the

initial two models for DLM, tissue:plasma PCs were calculated from ratios of areas

under tissue concentration versus time curves (AUCs) to the plasma AUC in oral dosing

Page 54: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

39

studies in rats (Mirfazaelian et al., 2006; Tornero-Velez et al., 2010). These

tissue:plasma PCs for DLM were subsequently adopted for modeling CIS and TRANS

(Tornero-Velez et al., 2012), due to a lack of PCs for PER. A computational approach

was adopted to estimate PCs for a third DLM model (Godin et al., 2010). A significant

problem with using these PC values is that the AUCs from in vivo single dose

toxicokinetic studies were not determined under steady-state conditions. Most of the

data used for estimating tissue:plasma PCs was obtained at relatively high doses

compared to environmental exposure. Because of the low analytical sensitivity of

pyrethroids, these high doses were required to obtain measurable tissue

concentrations. Modelers must assume that accurate analytical methods were carefully

performed by research groups. A likely, but frequently unrecognized problem with

analyses of highly-lipophilic compounds is their pronounced adherence, or non-specific

binding to, most plastic and glass devices. This was found to be true for deltamethrin

and permethrin, when measuring their plasma protein binding (Sethi et al., 2014).

A number of other approaches have been used as well with varying success to

determine tissue:plasma/blood PCs for pyrethroids and other highly lipophilic chemicals.

Some researchers have advocated the use of computational methods in the interest of

saving the time and cost of conducting animal studies. Poulin and Krishnan (Poulin and

Krishnan, 1995b) assumed the solubility of a chemical in n-octanol corresponded to the

sum total of its solubility in neutral lipids, phospholipids and water. The solubility of

highly lipophilic compounds in water was considered negligible, and solubility in

phospholipids 30% of that in neutral lipids. There was reasonable agreement between

their predicted and empirical PCs for 23 organic chemicals for some human tissues but

Page 55: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

40

not others. The predictions improved when solubility in olive or corn oil rather than n-

octanol was used as a surrogate for neutral liposolubility (Poulin and Krishnan, 1995a).

This approach worked well when it was utilized to calculate tissue:air and blood:air PCs

for volatile organic compounds (VOCs) (Poulin and Krishnan, 1996a; Poulin and

Krishnan, 1996b; Smith et al., 2005). Although tissue:blood PCs can readily be

calculated when tissue:air and blood:air PCs are available, this approach cannot be

used for pyrethroids, as their volatility is negligible. Payne and Kenny (Payne and

Kenny, 2002) assessed the relative accuracy, strengths and shortcomings of 10

published algorithms for estimating tissue:blood PCs. Accuracy was problematic in most

cases due to difficulties in modeling the effects of protein binding. DLM, CIS and

TRANS, like other very lipophilic compounds, are highly bound to plasma proteins and

lipoproteins (Sethi et al., 2014). More recently Krishnan and his colleagues (Peyret et

al., 2010) have published what is termed a unified algorithm by integrating additional

mechanistic algorithms, including estimations of hydrophobic interactions with neutral

lipids and binding to plasma proteins.

Estimation of tissue:blood PCs by in vitro techniques offers a relatively

inexpensive, time-saving alternative to in vivo approaches for non-volatile chemicals.

Reasonably good agreement between in vitro and in vivo PCs has been reported for

chemicals such as estradiol and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) (Murphy et

al., 1995). Estradiol and TCDD were each dissolved in propylene carbonate (PCARB)

and equilibrated for up to 5 hours with homogenized tissues and with blood.

Tissue:PCARB values divided by blood:PCARB values yielded tissue:blood PCs.

Reasonable agreement was observed for a series of barbituric acids, but the in vitro

Page 56: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

41

data underestimated in vivo tissue distribution for the most lipophilic congeners (Ballard

et al., 2003b). Albumin diffusion from tissue pieces to in vitro media was problematic for

highly plasma-bound compounds (Ballard et al., 2003a). CIS and TRANS are 80-90%

bound to albumin. The normal process of vascular perfusion of tissues does not occur in

in vitro systems. Homogenization enhances the penetration of test chemical, but results

in loss of structural integrity of cells and other potentially important cellular structures

such as the blood-brain barrier. A method for estimating non-volatile pesticides involving

ultrafiltration of tissue homogenates and pieces proved ineffective for compounds with a

Log P ≥ 1.7, due to retention by the filtration units employed (Tremblay et al., 2012). A

solid phase microextraction (SPME) technique was used for isolation of the more

lipophilic pesticides, but tissue:blood PCs could not be determined for compounds with

a Log P ˃ 4, due to excessive partitioning into the SPME fiber. Log P values of most

pyrethroids exceed 4. We found that adherence of pyrethroids to most glass, metals

and plastics to be a serious problem that could be minimized by using clean silanized

glass vials and LoBind® plastic pipettes (Sethi et al., 2014). In light of the uncertainties

with each approach, it is clear that pyrethroid PBPK models developed thus far, lack

reliable tissue:plasma PCs. Therefore, we decided to measure CIS and TRANS

tissue:plasma PCs in vivo at steady-state in order to have more assurance of the

reliability of input parameters for second generation PBPK models currently being

developed for these chemicals.

Although Alzet pumps® were widely utilized for the delivery of various

chemotherapeutic agents (Pun et al., 2004; Tamamura et al., 2003; Teicher et al.,

1992) in general, they have not seen a lot of attention for use in achieving steady-state.

Page 57: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

42

Alzet pumps® were used in the current study to maintain sustained delivery to reach

steady-state levels. Alzet pumps® allow us to determine tissue:plasma PCs at

environmentally relevant doses, which was a major drawback with the data reported

previously. All the pyrethroid PCs estimated so far were based on studies conducted in

adult rats. To our knowledge, no one has yet determined the tissue:plasma PCs of

pyrethroids in younger rats. Miniature Alzet pumps® were successfully employed in the

current study to achieve steady-state levels in younger rats.

The objectives of the current project were three-fold: (1) to develop an in vivo

model from which measurements of relative amounts of pyrethroids could be directly

determined at steady-state in adult rat tissues and plasma; (2) to utilize the model to

accurately determine tissue:plasma PCs for brain, liver, skeletal muscle, and fat for

DLM, CIS and TRANS; and (3) to determine whether these tissue:plasma PCs vary with

the stage of maturity of the test subjects.

MATERIALS AND METHODS

Chemicals and Materials

Cis-permethrin (CIS) (99.3%) and Trans-permethrin (TRANS) (99.0%) were kindly

provided by FMC Agricultural Products (Princeton, NJ). Deltamethrin (DLM) was

donated by Bayer Crop Science (Research Triangle Park, NC). Glycerol formal was

purchased from Acros Organics (NJ, USA). Acetonitrile (HPLC grade) and Spin-X®

microcentrifuge filters were purchased from Fisher Chemical Co. (Pittsburgh, PA).

Phosphoric acid (86%) was purchased from J.T. Baker (Phillipsburg, NJ). DNA Lo-bind®

microcentrifuge tubes were obtained from Eppendorf (Hauppauge, NY), and d-SPE

Page 58: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

43

(QuEChERS) kits were purchased from Agilent Technologies (Santa Clara, CA). Alzet®

osmotic (Model 2ML1) and micro-osmotic (Model 1007D) pumps were supplied by the

Durect Corporation (Cupertino, CA).

Animals

Adult male and timed-pregnant female Crl:CD SD rats were purchased from

Charles River Laboratories (Raleigh, NC). The protocol for this study was approved by

the University of Georgia Animal Care and Use Committee. Each rat was housed in a

cage with a 12-h light/12-h dark cycle at ambient temperature (22◦C) and relative

humidity (55 ± 5%). The females were allowed to deliver, and the pups housed with

their mothers until the offspring were 15 and 21 days old. The unsexed pups were

housed with their mother during CIS and TRANS exposure regimens. Food (5001

Rodent Diet, PMI Nutrition International LLC, Brentwood, MO, USA) and tap water were

provided ad libitum.

Adult and PND 21 rats were anesthetized using Ketamine, Acepromazine, and

Xylazine (KAX) cocktail @ 0.075 – 0.1 mL/100g body weight. Isoflurane was used to

anesthetize PND 15 rats due to KAX toxicity. The hair was trimmed at the site of

implantation on the dorsal surface of each unsexed pup and adult male rat. A small

incision was made, and a large subcutaneous (sc) pocket created by dissecting the

subcutaneous tissue. A DLM, CIS- or TRANS-loaded Alzet® pump was inserted into the

pocket with the delivery port first. The incision was then closed using wound clips, and

the rats returned to their cage. Animals were provided with food and water ad libitum

during recovery and pyrethroid infusion.

Page 59: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

44

In vitro Plasma:Blood Partition Coefficient Determination

Lo-bind® centrifuge tubes containing 100 µL of fresh control rat blood and 10 µL of

0.64 M sodium fluoride (NaF) were spiked with 5 µg DLM, CIS or TRANS and vortexed

for 10 sec (Mini Vortexer®, VWR) (West Chester, PA). The tubes were incubated for 10,

30, 45 and 60 min in an orbital shaker at 37°C. The tubes were then centrifuged for 5

min at 4,342×g to separate plasma and red blood cells (RBC). DLM, CIS and TRANS in

plasma and RBC were extracted and quantified using a modified high performance

liquid chromatography (HPLC) method developed by Kim et al (Kim et al., 2006). Briefly,

50 µL of separated plasma were placed into a Lo-bind® tube and spiked with 8 µg of

internal standard (DLM was used as internal standard for CIS and TRANS, and CIS was

used as internal standard for DLM), and vortexed for 40 sec. Seventy-five µL of

acetonitrile (ACN) were added and the resulting solution vortexed for 70 sec to denature

the plasma proteins. The contents of the centrifuge tube were then centrifuged for 10

min at 15,871×g, and 100 µL of supernatant was transferred to a vial for HPLC analysis.

The red blood cells (RBC) pellet was also spiked with 8 µg of DLM as an internal

standard and vortexed for 40 sec. One hundred µL of cold distilled water were added

and the suspension vortexed for 70 sec to lyse the RBC. Two hundred µL of ACN were

added, and the contents were vortexed for 70 sec to denature the proteins. The tubes

were then centrifuged at 15,871×g, and 150 µL of supernatant was taken for HPLC

analysis. The analysis was performed using an Agilent 1100 series HPLC (Foster City,

CA). The chromatographic separation was achieved using a Phenomenex® Luna C18

column. The mobile phase was methanol:HPLC water (97:3, v/v) operated at a constant

flow rate of 1 mL/min. Concentration in blood was calculated as the sum total of

Page 60: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

45

concentration in RBC and concentration in plasma. Plasma:blood PCs were calculated

by dividing the concentration of DLM, CIS or TRANS measured in the plasma by that

calculated in the blood.

In vivo Tissue:Plasma Partition Coefficient Measurement

An oral loading dose coupled with constant infusion of DLM, CIS or TRANS was

employed, in order to achieve systemic steady-state or equilibrium of the chemical in

adult and PND pups. Only constant infusion, however, was employed in PND 15 pups,

due to toxicity seen with combined loading and infusion doses.

An Alzet® infusion pump containing DLM, CIS or TRANS was surgically implanted

subcutaneously on adult rats’ dorsum, for constant infusions of 0.36 mg/h. Three to 4-h

later, each adult was gavaged with a “loading dose” of 30 mg/kg of DLM, or 150 mg/kg

of CIS or TRANS. After 72-h the adult rats were sacrificed by cervical dislocation and

blood withdrawn by closed-chest cardiac puncture into a heparinized syringe. Whole

blood was centrifuged immediately to separate plasma. Ten µL of 0.64M NaF were

added for every 100 µL of plasma collected to inhibit esterases. The abdominal cavity

was opened and sterile saline injected into the portal vein, in order to flush even more

blood from the liver and other organs. Samples of whole brain, liver, thigh muscle and

perirenal fat were excised, immediately flash-frozen with liquid nitrogen and stored at -

80°C until analysis.

PND 15 and 21 pups with subcutaneous Alzet® pumps were infused with DLM or

CIS at rates of 0.03 mg/h and 0.02 mg/h, respectively. PND 15 and 21 pups were

infused with 0.05 mg TRANS/h. PND 21 pups were also administered a loading dose of

Page 61: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

46

0.25 mg/kg DLM or CIS, or 1.15 mg/kg TRANS by gavage 3-4 h after the start of

infusion. PND 15 pups did not receive a loading dose. After 48 and 72-h of infusion,

groups of the pups were sacrificed by cervical dislocation. The maximum amount of

blood possible was withdrawn by closed-chest cardiac puncture. Plasma was separated

from whole blood as described above, and samples of whole brain, liver and abdominal

muscle were excised and flash frozen like tissues from adults. Fat was not harvested

from PND 15 and 21 pups due to lack of adipose tissue.

Tissue Extraction and Analysis

Brain and liver were homogenized in 2× volume of saline, while muscle and fat

were homogenized in 3× volume of saline and water respectively, using a Tekmar

Tissumizer® (Cincinnati, OH). Plasma and fat homogenates were extracted using a

method developed by Gullick et al (Gullick et al., 2014). Briefly, 100 µL of plasma or fat

homogenate were extracted with 500 µL of an extracting solution comprised of internal

standard (10 ng/mL DLM for CIS and TRANS analyses, and 50 ng/mL CIS for DLM

analyses) in a mixture of 1% phosphoric acid and 99% ACN. The sample was mixed

and centrifuged before drying the solvent and reconstitution with 6 µl toluene for

analysis. Brain, muscle and liver samples were extracted using an improved

bioanalytical method also developed by Gullick et al (Gullick et al., 2016). Three

hundred µl of tissue homogenate were extracted with 800 µl of an extracting solution

comprised of internal standard (10 ng/mL DLM for CIS and TRANS analyses, and 50

ng/mL CIS for DLM analyses) in n-hexane-saturated acetonitrile. The sample was

mixed and centrifuged. Sample clean-up of the supernatant was performed using

Page 62: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

47

QuEChERS microcentrifuge tubes before drying the solvent and reconstitution with 6 µL

of toluene for analysis by GC-negative chemical ionization-MS (GC-NCI-MS).

GC-NCI-MS analysis was performed using an Agilent 6890N gas chromatograph

with a 5973 quadrupole mass selective detector and Enhanced Chemstation (Build 75

Agilent, 2003). Chromatographic separation was achieved using a Zebron® ZB5-MS GC

column operating at 1 mL/min constant flow helium.

Determination of Partition Coefficients

For each animal, the steady-state concentration of DLM, CIS or TRANS in each tissue

being studied was divided by the steady-state concentration in plasma, in order to

determine the tissue:plasma PC. Means and standard deviations (SDs) were calculated

with Microsoft Excel (Microsoft, Redmond, WA). All experiments were performed with a

minimum of 4 animals unless otherwise stated. Statistical significance was evaluated

between groups using either the Student’s t-test or one way ANOVA, followed by

Tukey’s multiple comparison test, as indicated, with a significance level of p<0.05, using

Graphpad Prism 6 software® (San Diego, CA).

RESULTS

Partitioning of DLM, CIS and TRANS between Plasma and Blood

The concentration of each compound in plasma and RBC was measured periodically by

HPLC during 1-h incubation (Table 2.1). It is evident that the concentration of CIS and

TRANS progressively decreased in the RBC and increased in the plasma during the 60

min incubation period, with the most pronounced changes occurring during the initial 30

Page 63: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

48

min. Concentration of DLM in the RBC and plasma did not change over the incubation

period. Concentrations of each compound are ~3 fold higher in plasma than RBC at 60

min. If it is assumed that plasma and RBCs account for 60% and 40% of the volume of

rat blood, respectively, ~80% of each compound is present in the plasma.

Partitioning of DLM, CIS and TRANS between Plasma and Tissues of Adult Rats

Plasma concentrations of DLM, CIS and TRANS were measured periodically in

adult rats in pilot experiments to determine when steady-state was attained. It is

generally accepted that steady-state will be reached in ≤ 5 half-lives of a chemical.

Since Anandon et al. (Anadón et al., 1991) reported the half-life of permethrin in adult

SD rats to be 12.4-h, steady-state should be achieved in approximately 62-h. We

observed considerable intersubject variability in plasma CIS levels at 24 and 48-h, so it

was decided to sacrifice adult animals after 72-h of the infusion protocol for plasma and

tissue analyses. Mean plasma TRANS levels at 48 and 72-h were comparable, so the

animals’ sacrifice for the tissue:plasma TRANS PC determination was also performed

after 72-h. Similarly, the half-life (t½) of DLM, when given to adult SD rats in glycerol

formal (GF) by gavage, was reported by Kim et al. (2008) to be 13.3- h, and the steady-

state was expected to be achieved in approximately 67-h. Similar to TRANS, DLM

mean plasma levels were comparable at 48 and 72-h. So the animals were sacrificed

after 72-h of constant infusion.

CIS and TRANS exhibited the same pattern of tissue distribution and the same rank

order of tissue:plasma PCs in adult rats (Table 2.2B & 2.2C). The CIS liver:plasma PC

was lowest, followed in ascending order by the brain, skeletal muscle and fat. TRANS

Page 64: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

49

PCs showed the same rank order, although TRANS levels in the liver of many subjects

were below the limit of quantitation (LOQ). The fat:plasma PC for each isomer was far

higher than that for the other tissues analyzed. The brain:plasma PCs for CIS and

TRANS were quite similar. The CIS fat:plasma PCs, however, were about 1.5-fold

higher than the TRANS PCs.

For DLM, the brain:plasma PC was lowest, followed by liver, skeletal muscle and

fat (Table 2.2A). The brain:plasma and fat:plasma PCs for DLM were about 2 - 3 fold

lower when compared to CIS and TRANS. However, DLM liver:plasma and

muscle:plasma PCs were higher than that of CIS and TRANS. Similar to CIS and

TRANS, after 72-h of constant infusion, fat accumulated far higher levels of DLM than

other tissues.

Partitioning of DLM, CIS and TRANS between Plasma and Tissues of Pups

The duration of infusions of PND 15 and 21 rats did not substantially influence

most tissue:plasma PCs. In both PND 21 and PND 15 pups, the 48 and 72-h

tissue:plasma PCs for DLM were similar (Table 2.4A). In PND 21 pups, the 72-h

brain:plasma PC for CIS was higher than that determined at 48-h (Table 2.4B).

Although the 72-h muscle:plasma PC was also higher than that of 48-h, this difference

failed to reach statistical significance. However, there was no change in liver:plasma PC

at 48 and 72-h. The 72-h plasma TRANS concentrations in PND 21 pups were about

half of that at 48 h, as were the tissue concentrations (Table 2.4C). The TRANS

brain:plasma and muscle:plasma PCs remained about the same. Plasma, brain and

liver concentrations of CIS remained relatively unchanged from 48 to 72-h in the PND

Page 65: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

50

15 pups (Table 2.5B). Accordingly, the PCs remained much the same. Plasma, brain

and liver TRANS levels decreased modestly in parallel, resulting in equivalent or

comparable brain:plasma and liver:plasma PCs at 48 and 72-h (Table 2.5C).

Nevertheless, skeletal muscle exhibited increased partitioning of CIS and TRANS

between 48 and 72-h, with an ensuing increase in the muscle:plasma PC for both

isomers. This is in contrast to DLM, where muscle:plasma PCs remained almost the

same between 48 and 72-h (Table 2.5A).

In most cases, the results of the current study indicate that DLM, CIS and

TRANS tissue:plasma PCs in younger pups were higher as compared to adult rats.

While the brain:plasma and liver:plasma PCs for DLM in PND 15 pups were significantly

higher as compared to adults, the muscle:plasma PC was not significantly different.

Surprisingly, tissue:plasma PCs for DLM in PND 21 pups were consistently lower as

compared to PND 15 pups and adults (Fig 2.2). Tissue:plasma PCs for DLM in PND 15

and PND 21 pups were consistently lower as compared to CIS and TRANS. CIS

brain:plasma, liver:plasma and muscle:plasma PCs in PND 15 and PND 21 pups were

significantly higher than in adults (Fig 2.3). Although the liver:plasma PC in PND 21 was

significantly lower than PND 15 pups, no significant differences were observed in the

brain:plasma and muscle:plasma PCs between PND 15 and PND 21 pups (Fig 2.3).

The TRANS muscle:plasma PCs and likely the liver:plasma PCs were inversely

proportional to age (Fig 2.4). The TRANS concentrations in the liver of PND 21 pups

were below the LOQ and could not be measured. Interestingly, the PND 21

muscle:plasma PC did not differ significantly from that of adults. Unexpectedly, all three

age-groups exhibited comparable TRANS brain:plasma PCs. It was not possible to

Page 66: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

51

assess the influence of maturation on fat:plasma PCs, as there was no visible adipose

tissue in the PND 15 or 21 pups.

DISCUSSION

Accurate tissue:plasma PCs are essential input parameters for PBPK models, as

PCs describe the distribution of a test compound in key tissues of the body. There is a

lack of reliable methods for the measurement of tissue:plasma PCs at steady-state.

None of the PC values reported previously for pyrethroids were measured at steady-

state. Also, relatively high doses were often employed due to the low analytical

sensitivity of pyrethroids. These high doses are not environmentally relevant and may

saturate the distribution and elimination processes. No one has yet established whether

tissue distribution and PCs of pyrethroids vary significantly during maturation and

development. In the current study, we successfully used Alzet pumps® to attain steady-

state and accurately determine tissue:plasma PCs both in adult and younger rats.

The fat:plasma PCs of 45, 98 and 67, determined in the current investigation for

DLM, CIS and TRANS respectively, were far higher than for other tissues monitored in

adult rats. Tissues with a sizable fat content would be expected to accumulate the

largest amounts of highly lipophilic chemicals such as pyrethroids, and thus exhibit

relatively high tissue:plasma PCs. Rat adipose tissue is comprised of ~85% lipids,

13.5% extracellular water and 1.5% intracellular water (Rodgers et al., 2005). About

99.7% of the lipids are neutral lipids (Schmitt, 2008). Skeletal muscle contains just 2%

lipids (Rodgers et al., 2005), but exhibited significantly higher DLM, CIS and TRANS

PCs than brain or liver. This may be attributable to interspersed fat in the specimens of

Page 67: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

52

muscle used for the determination. Unexpectedly, levels of DLM, CIS and TRANS in

brain were similar to or lower than levels in plasma. It was previously reported that the

DLM levels in brain were substantially lower than in blood (Kim et al., 2008; Mirfazaelian

et al., 2006). Rat brain contains 4% lipids, of which more than half are composed of

neutral and acidic phospholipids. The solubility of lipophilic chemicals in phospholipids

is assumed to be just 30% of that in neutral lipids (Poulin and Krishnan, 1995b). The

total lipid content of liver is even lower and its proportion of phospholipids higher than in

brain (Schmitt, 2008). The liver of adult and PND 21 rats exhibited the lowest of any CIS

PCs. Concentrations of TRANS in the liver of both age-groups were below the LOQ.

Willemin et al. (Willemin et al., 2016) monitored the time-course of CIS and TRANS, but

were also unable to quantify TRANS in the liver, kidney or testes of adult, male SD rats

gavaged with 25 mg/kg of each isomer. Previous toxicokinetic studies with pyrethroids

indicated that adipose tissue accumulates the highest levels of DLM, CIS and TRANS,

followed in order by skin, skeletal muscle and other organs (Kim et al., 2008; Tornero-

Velez et al., 2012).

Tissue:plasma DLM, CIS and TRANS PCs in adult rats obtained in the current

steady-state study were compared in Table 2.3 with tissue:blood PCs derived from the

pharmacokinetic data of other investigators. Those PC values were calculated by

dividing each research group’s published tissue AUCs by the corresponding

blood/plasma AUCs. The DLM tissue:plasma PCs obtained from the current study were

quite comparable to those previously reported. This can be attributed to the use of the

same experimental approach. Both Mirfazaelian et al. and Kim et al. used male SD rats

as test species and glycerol formal as vehicle to dissolve DLM. The pattern of

Page 68: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

53

tissue:plasma PCs for CIS and TRANS obtained in the current project is also similar to

those estimated by other investigators, but some differences should be noted. The

differences are likely attributable, at least in part, to use of different doses and

approaches to derive the values. Tornero-Velez et al. (Tornero-Velez et al., 2012)

monitored blood and tissue CIS and TRANS concentrations in adult Long Evans rats

sacrificed periodically after being given 1 or 10 mg/kg of a CIS/TRANS (40:60) mixture

by corn oil gavage. Willemin et al. (Willemin et al., 2016) measured tissue and blood

levels of CIS and TRANS in adult SD rats given 25 mg/kg of each isomer by corn oil

gavage. These CIS brain:blood PCs are higher than the currently reported steady-state

brain:plasma PC. The TRANS steady-state brain:plasma PC of 0.7 is about the same

as the published PCs, with the exception of the inordinately high 1 mg/kg value. The

CIS steady-state liver:plasma PC is somewhat lower than its previously reported

counterparts. It should be remembered that, in general, blood concentrations will be

lower as compared to plasma, due to its larger volume. Using AUCs from blood

concentration time profiles may over estimate PCs, which might also be a reason for the

higher PC values reported previously. CIS steady-state fat:plasma PC is quite

comparable to the fat:blood PCs of Tornero-Velez et al. (Tornero-Velez et al., 2012), but

lower than that of Willemin et al. (Willemin et al., 2016) Interestingly, all four TRANS

fat:plasma/blood PCs are very similar.

Although, PCs are commonly assumed to be independent of dose, saturation of

plasma binding and clearance mechanisms by high concentrations of pyrethroids may

significantly alter their partitioning. Beliveau and Krishnan (Krishnan, 2000) observed

increase in the unbound fraction (fu) with increase in the blood concentration of four

Page 69: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

54

volatile organic chemicals (VOCs). Protein binding constants typically rise in concert

with chemicals’ lipophilicity. CIS and TRANS are highly lipophilic (permethrin Log P =

6.3), bind hydrophobically to albumin and lipoprotein, and are 90% bound to human

plasma at toxicologically-relevant concentrations (Sethi et al., 2014). Plasma binding

becomes saturated when CIS and TRANS concentrations approach and exceed 300

ng/ml. Blood TRANS levels did not exceed 300 ng/ml in the in vivo experiments of

Willemin et al. (Willemin et al., 2016) or Tornero-Velez et al., (Tornero-Velez et al.,

2012) or in the current steady-state experiment. The TRANS brain:blood and fat:blood

PCs based on these published data, with the exception of the 1 mg/kg value of Tornero-

Velez et al., are similar to one another and to the corresponding steady-state TRANS

PCs for adult rats (Table 3). The Willemin et al. CIS brain:blood, liver:blood and

fat:blood PCs, however, are generally higher than the respective Tornero-Velez et al.

PCs and our steady-state PCs. This may be attributable to Willemin et al’s. CIS blood

levels of over 1,000 ng/mL, which would have exceeded the binding capacity of plasma.

This blood level of 1,000 ng/mL CIS is much higher when compared to Tornero-Velez et

al.’s 342 ng/mL and 32 ng/mL in the current study. Another important factor that could

account for differences in the PCs between these studies is the lack of steady-state in

the toxicokinetic studies. Time-course profiles for DLM, CIS and TRANS in brain and fat

differ from those for blood, liver and kidney, reflecting differences in rates of tissue

uptake and clearance. It is known that plasma binding of the pyrethroids is

concentration-dependent and can govern their rate of tissue uptake (Amaraneni et al.,

2016). The blood-brain barrier serves to limit and delay DLM uptake into the adult rat’s

brain (Amaraneni et al., 2016). At steady-state, PCs are independent of the kinetics of

Page 70: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

55

distribution into and clearance from tissues. Once steady-state is attained, the plasma fu

is stable, allowing a constant rate of input into tissues. At steady-state, the plasma and

tissue compartments are in equilibrium, with the exception of fat which continues to

accumulate the highly lipophilic pyrethroids. Clearance will also be constant at a single

steady-state concentration. Clearance in vivo would be expected to vary with

concentration and become saturated due to the dose-dependent processes (plasma

protein binding, metabolism) characteristic of pyrethroids.

To assess the influence of maturation on the distribution of pyrethroids, we

measured the tissue:plasma PCs of DLM, CIS and TRANS in PND 15 and PND 21

pups. Tissue:plasma PCs for DLM were highest in the PND 15 rats as compared to both

PND 21 and adult rats. The lower capacity of metabolic enzymes combined with lower

plasma binding capacity might have resulted in greater distribution of DLM to the brain

and liver. However, the muscle:plasma PC in this age group is lower than adults. This

might be attributable to the lower fat content in muscle tissue in this age group.

Surprisingly, tissue:plasma PCs in PND 21 pups were consistently lower when

compared to PND 15 and adult rats. DLM in rats is rapidly metabolized by both

hydrolysis by CEs and oxidation by CYP 1A1 and 3A2 (Godin et al., 2007). De Zwart et

al. (de Zwart et al., 2008) reported that plasma and liver CE activity, and CYP 1A1 and

3A2 activity in SD rats reach adult levels by PND 26. The lower tissue:plasma PCs

found in PND 21 pups might be a result of rapid metabolism of lower doses of DLM (LD

of 0.25 mg/kg vs 30 mg/kg in adults) in this age group. Sethi et al. (Sethi et al., 2015)

reported that the total plasma protein binding of DLM in PND 21 pups is not significantly

different from that of adult rats. Rapid metabolism combined with increased plasma

Page 71: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

56

protein binding might have contributed to lower tissue:plasma PCs for DLM in PND 21

pups.

Tissue:plasma PCs for permethrin’s isomers, notably CIS, generally decreased

during maturation. Tissue concentrations of each isomer were consistently highest in

the PND 15 rats in the current study. Elevated liver CIS levels in immature animals likely

reflect diminished capacity to metabolize the pyrethroid and reduced ability to retain it in

the bloodstream due to relatively low plasma protein levels. There are apparently no

data on the age-dependence of permethrin biotransformation, although Anand et al.

(Anand et al., 2006) described a progressive marked increase in CYP P450- and CE-

mediated hepatic metabolism of DLM between PND 10 and 90 in SD rats. There was

also a doubling of the total plasma protein level in SD rats between PND 10 and 60.

Blood:air PCs for four VOCs were reported to be higher in mature than in immature rats

(Mahle et al., 2007). Lerman et al. (Lerman et al., 1984) found a direct correlation

between serum albumin and triglyceride levels in neonates, children and adults and

blood:air PCs of a series of lipophilic anesthetics. Lower capacity of the plasma of

immature humans and rodents to bind and retain lipophilic compounds undoubtedly

contributes to higher liver levels and liver:plasma PCs. This situation should be

compounded by elevation of systemic levels of lipophiles as a result of substantially

reduced body fat stores in immature subjects. Unfortunately liver TRANS levels were

too low in our steady-state experiment and in the in vivo study of Willemin et al.

(Willemin et al., 2016) to calculate TRANS liver:plasma PCs for adult or PND 21 rats.

Rapid metabolism of TRANS by CE mediated hydrolysis was responsible for levels

below the LOQ.

Page 72: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

57

Steady-state brain:plasma PCs measured for permethrin’s (PER’s) isomers did

not exhibit a consistent pattern of change during rats’ maturation. The CIS brain:plasma

PCs appeared to gradually increase with age, while the TRANS brain:plasma PC was

essentially unchanged. One explanation for this dichotomy might be the presence of

relatively high plasma CIS levels that saturate plasma binding in pups, resulting in

greater diffusion from the blood to brain and other tissues. This may also account for the

relatively high CIS muscle:plasma PCs we observed in pups. Greater permeability of

the blood-brain barrier to pyrethroids could also contribute to elevated brain deposition

and a higher brain:plasma PC in the immature animals. Conversely, increased

myelination during development should tend to enhance CNS deposition of pyrethroids.

An increase of 35% in brain lipid content has been measured in S-D rats between PND

16 and 24 (Galli and Cecconi, 1967). This is reflected in reported increases in brain:air

PCs from birth to adulthood for lipophilic anesthetic gases in humans (Lerman et al.,

1986) and VOCs in rats (Krishnan, 2000).

In Table 2.6, steady-state tissue:plasma PCs obtained from the current study in

PND 15, PND 21 and adult rats were compared to those estimated from AUCs obtained

from a single dose toxicokinetic study (Tanzir et al., Chen et al., to be submitted). We

found that while some tissue:plasma PCs were comparable to each other, some were

markedly different. It should be remembered that there were some differences among

the doses and vehicles used in both the studies. Steady-state tissue:plasma PCs for

DLM obtained from the current study were quite similar to those estimated from AUCs,

especially in younger age groups (Table 2.6A). Steady-state plasma levels of 78 and 10

ng/mL in PND 21 and PND 15 pups respectively in the current study matched well with

Page 73: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

58

the respective peak plasma concentrations of 47 and 21 ng/mL in the single dose study.

This might probably be the reason for comparable tissue:plasma PCs obtained from

both approaches. For CIS and TRANS, while steady-state brain:plasma PCs were

comparable to those estimated from AUCs, steady-state muscle:plasma and fat:plasma

PCs were somewhat lower than their counterparts (Tables 2.6 B&C). Steady-state

liver:plasma PCs were quite lower comparted to those estimated from AUCs. The peak

plasma concentrations for CIS and TRANS (5 – 8 µg/mL) from single dose studies (600

mg/kg) were significantly higher than the respective steady-state plasma levels (30 –

80 ng/mL). Pronounced age-dependent differences in the tissue:plasma PCs estimated

from AUCs were only evident in the liver tissue. Saturation of plasma protein binding

and clearance mechanisms at high doses employed in the single dose study might be

the reason for not seeing pronounced age-dependent differences in the tissue:plasma

PCs estimated from AUCs.

In conclusion, we have developed a means of achieving steady-state for highly

lipophilic chemicals in a rodent, in order to measure tissue:plasma PCs as accurately as

possible in vivo. Our results indicate that the adipose tissue was the major repository for

DLM, CIS and TRANS in the rat, followed by skeletal muscle, after 72-h of constant

infusion. As a result, the tissue:plasma PC for fat was higher than for other tissues.

Brain:plasma and liver:plasma PCs were lower than unity in adult animals. The liver

concentrations of TRANS were below LOQ in adult and PND 21 rats, apparently due to

rapid hydrolysis by carboxylesterases and oxidation by cytochrome P450s.

Tissue:plasma PCs were found to be inversely proportional to age and decreased with

increasing age. These differences become more pronounced as the age diminishes. A

Page 74: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

59

few exceptions were observed in the PND 21 rats. It was not possible to assess the

influence of maturity on partitioning of pyrethroids into fat, due to lack of visible adipose

tissue in younger rats. The knowledge from these studies can be used by other

researchers in the future to understand the disposition of other pesticides which have

similar physicochemical properties.

Page 75: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

60

REFERENCES

Amaraneni, M., Sharma, A., Pang, J., Muralidhara, S., Cummings, B. S., White, C. A.,

Bruckner, J. V., and Zastre, J. (2016). Plasma protein binding limits the blood brain

barrier permeation of the pyrethroid insecticide, deltamethrin. Toxicology letters 250, 21-

28.

Anadón, A., Martinez-Larranaga, M., Diaz, M., and Bringas, P. (1991). Toxicokinetics of

permethrin in the rat. Toxicology and applied pharmacology 110(1), 1-8.

Anadon, A., Martinez-Larranaga, M., Fernandez-Cruz, M., Diaz, M., Fernandez, M., and

Martinez, M. (1996). Toxicokinetics of deltamethrin and its 4′-HO-metabolite in the rat.

Toxicology and applied pharmacology 141(1), 8-16.

Anand, S. S., Kim, K.-B., Padilla, S., Muralidhara, S., Kim, H. J., Fisher, J. W., and

Bruckner, J. V. (2006). Ontogeny of hepatic and plasma metabolism of deltamethrin in

vitro: role in age-dependent acute neurotoxicity. Drug metabolism and disposition 34(3),

389-397.

Andersen, M. E. (2003). Toxicokinetic modeling and its applications in chemical risk

assessment. Toxicology letters 138(1), 9-27.

Ballard, P., Arundel, P. A., Leahy, D. E., and Rowland, M. (2003a). Prediction of in vivo

tissue distribution from in vitro data. 2. Influence of albumin diffusion from tissue pieces

Page 76: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

61

during an in vitro incubation on estimated tissue-to-unbound plasma partition

coefficients (kpu). Pharmaceutical research 20(6), 857-863.

Ballard, P., Leahy, D. E., and Rowland, M. (2003b). Prediction of in vivo tissue

distribution from in vitro data. 2. Correlation between in vitro and in vivo tissue

distribution of a homologous series of nine 5-n-alkyl-5-ethyl barbituric acids.

Pharmaceutical research 20(6), 864-872.

Barr, D. B., Olsson, A. O., Wong, L.-Y., Udunka, S., Baker, S. E., Whitehead Jr, R. D.,

Magsumbol, M. S., Williams, B. L., and Needham, L. L. (2010). Urinary concentrations

of metabolites of pyrethroid insecticides in the general US population: National Health

and Nutrition Examination Survey 1999-2002. Environmental Health Perspectives

118(6), 742.

de Zwart, L., Scholten, M., Monbaliu, J. G., Annaert, P. P., Van Houdt, J. M., Van den

Wyngaert, I., De Schaepdrijver, L. M., Bailey, G. P., Coogan, T. P., and Coussement,

W. C. (2008). The ontogeny of drug metabolizing enzymes and transporters in the rat.

Reproductive Toxicology 26(3), 220-230.

Galli, C., and Cecconi, D. R. (1967). Lipid changes in rat brain during maturation. Lipids

2(1), 76-82.

Page 77: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

62

Godin, S. J., Crow, J. A., Scollon, E. J., Hughes, M. F., DeVito, M. J., and Ross, M. K.

(2007). Identification of rat and human cytochrome p450 isoforms and a rat serum

esterase that metabolize the pyrethroid insecticides deltamethrin and esfenvalerate.

Drug Metabolism and Disposition 35(9), 1664-1671.

Godin, S. J., DeVito, M. J., Hughes, M. F., Ross, D. G., Scollon, E. J., Starr, J. M.,

Setzer, R. W., Conolly, R. B., and Tornero-Velez, R. (2010). Physiologically based

pharmacokinetic modeling of deltamethrin: development of a rat and human diffusion-

limited model. Toxicological Sciences doi, kfq051.

Godin, S. J., Scollon, E. J., Hughes, M. F., Potter, P. M., DeVito, M. J., and Ross, M. K.

(2006). Species differences in the in vitro metabolism of deltamethrin and esfenvalerate:

differential oxidative and hydrolytic metabolism by humans and rats. Drug metabolism

and disposition 34(10), 1764-1771.

Gullick, D., Popovici, A., Young, H. C., Bruckner, J. V., Cummings, B. S., Li, P., and

Bartlett, M. G. (2014). Determination of deltamethrin in rat plasma and brain using gas

chromatography–negative chemical ionization mass spectrometry. Journal of

Chromatography B 960, 158-165.

Gullick, D. R., Bruckner, J. V., White, C. A., Chen, C., Cummings, B. S., and Bartlett, M.

G. (2016). Quantitation of Deltamethrin in Rat Liver and Muscle Homogenates Using

Page 78: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

63

Dispersive Solid-Phase Extraction with GC-NCI-MS. Journal of AOAC International

99(3), 813-820.

Kim, K.-B., Anand, S. S., Kim, H. J., White, C. A., and Bruckner, J. V. (2008).

Toxicokinetics and tissue distribution of deltamethrin in adult Sprague–Dawley rats.

Toxicological sciences 101(2), 197-205.

Kim, K. B., Bartlett, M. G., Anand, S. S., Bruckner, J. V., and Kim, H. J. (2006). Rapid

determination of the synthetic pyrethroid insecticide, deltamethrin, in rat plasma and

tissues by HPLC. Journal of Chromatography B 834(1), 141-148.

Krishnan, M. B., Kannan (2000). Concentration dependency of rat blood: air partition

coefficients of some volatile organic chemicals. Journal of Toxicology and

Environmental Health Part A 60(6), 377-389.

Lerman, J., Gregory, G., Willis, M., and Eger 2nd, E. (1984). Age and solubility of

volatile anesthetics in blood. Anesthesiology 61(2), 139-143.

Lerman, J., Schmitt-Bantel, B., Gregory, G., Willis, M., and Eger 2nd, E. (1986). Effect

of age on the solubility of volatile anesthetics in human tissues. Anesthesiology 65(3),

307-311.

Page 79: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

64

Lipscomb, J. C., Haddad, S., Poet, T., and Krishnan, K. (2012). Physiologically-based

pharmacokinetic (PBPK) models in toxicity testing and risk assessment. In New

Technologies for Toxicity Testing (doi, pp. 76-95. Springer.

Lu, C., Barr, D. B., Pearson, M., Bartell, S., and Bravo, R. (2006). A longitudinal

approach to assessing urban and suburban children's exposure to pyrethroid pesticides.

Environmental health perspectives doi, 1419-1423.

Mahle, D. A., Gearhart, J. M., Grigsby, C. C., Mattie, D. R., Barton, H. A., Lipscomb, J.

C., and Cook, R. S. (2007). Age-dependent partition coefficients for a mixture of volatile

organic solvents in Sprague-Dawley rats and humans. Journal of Toxicology and

Environmental Health, Part A 70(20), 1745-1751.

Mirfazaelian, A., Kim, K.-B., Anand, S. S., Kim, H. J., Tornero-Velez, R., Bruckner, J. V.,

and Fisher, J. W. (2006). Development of a physiologically based pharmacokinetic

model for deltamethrin in the adult male Sprague-Dawley rat. Toxicological Sciences

93(2), 432-442.

Morgan, M. K. (2012). Children’s exposures to pyrethroid insecticides at home: a review

of data collected in published exposure measurement studies conducted in the United

States. International journal of environmental research and public health 9(8), 2964-

2985.

Page 80: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

65

Murphy, J. E., Janszen, D. B., and Gargas, M. L. (1995). An in vitro method for

determination of tissue partition coefficients of non‐volatile chemicals such as 2, 3, 7, 8‐

tetrachlorodibenzo‐p‐dioxin and estradiol. Journal of Applied Toxicology 15(2), 147-152.

Payne, M. P., and Kenny, L. C. (2002). Comparison of models for the estimation of

biological partition coefficients. Journal of Toxicology and Environmental Health Part A

65(13), 897-931.

Peyret, T., Poulin, P., and Krishnan, K. (2010). A unified algorithm for predicting

partition coefficients for PBPK modeling of drugs and environmental chemicals.

Toxicology and applied pharmacology 249(3), 197-207.

Poulin, P., and Krishnan, K. (1995a). An algorithm for predicting tissue: blood partition

coefficients of organic chemicals from n‐octanol: water partition coefficient data. Journal

of Toxicology and Environmental Health, Part A Current Issues 46(1), 117-129.

Poulin, P., and Krishnan, K. (1995b). A biologically-based algorithm for predicting

human tissue: blood partition coefficients of organic chemicals. Human & experimental

toxicology 14(3), 273-280.

Poulin, P., and Krishnan, K. (1996a). A mechanistic algorithm for predicting blood: air

partition coefficients of organic chemicals with the consideration of reversible binding in

hemoglobin. Toxicology and applied pharmacology 136(1), 131-137.

Page 81: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

66

Poulin, P., and Krishnan, K. (1996b). A tissue composition-based algorithm for

predicting tissue: air partition coefficients of organic chemicals. Toxicology and applied

pharmacology 136(1), 126-130.

Pun, S. H., Tack, F., Bellocq, N. C., Cheng, J., Grubbs, B. H., Jensen, G. S., Davis, M.

E., Brewster, M., Janicot, M., and Janssens, B. (2004). Targeted delivery of RNA-

cleaving DNA enzyme (DNAzyme) to tumor tissue by transferrin-modified, cyclodextrin-

based particles. Cancer biology & therapy 3(7), 641-650.

Rodgers, T., Leahy, D., and Rowland, M. (2005). Physiologically based pharmacokinetic

modeling 1: Predicting the tissue distribution of moderate‐to‐strong bases. Journal of

pharmaceutical sciences 94(6), 1259-1276.

Rowland, M., Peck, C., and Tucker, G. (2011). Physiologically-based pharmacokinetics

in drug development and regulatory science. Annual review of pharmacology and

toxicology 51, 45-73.

Schmitt, W. (2008). General approach for the calculation of tissue to plasma partition

coefficients. Toxicology in Vitro 22(2), 457-467.

Sethi, P. K., Muralidhara, S., Bruckner, J. V., and White, C. A. (2014). Measurement of

plasma protein and lipoprotein binding of pyrethroids. Journal of pharmacological and

toxicological methods 70(1), 106-111.

Page 82: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

67

Sethi, P. K., White, C. A., Cummings, B. S., Hines, R. N., Muralidhara, S., and

Bruckner, J. V. (2015). Ontogeny of Plasma Proteins, Albumin and Binding of

Diazepam, Cyclosporine and Deltamethrin. Pediatric research doi.

Shardlow, C. E., Generaux, G. T., Patel, A. H., Tai, G., Tran, T., and Bloomer, J. C.

(2013). Impact of physiologically based pharmacokinetic modeling and simulation in

drug development. Drug Metabolism and Disposition 41(12), 1994-2003.

Smith, A., Campbell, J., Keys, D., and Fisher, J. (2005). Rat tissue and blood partition

coefficients for n-alkanes (C8 to C12). International journal of toxicology 24(1), 35-41.

Soderlund, D. M. (2012). Molecular mechanisms of pyrethroid insecticide neurotoxicity:

recent advances. Archives of toxicology 86(2), 165-181.

Tamamura, H., Hori, A., Kanzaki, N., Hiramatsu, K., Mizumoto, M., Nakashima, H.,

Yamamoto, N., Otaka, A., and Fujii, N. (2003). T140 analogs as CXCR4 antagonists

identified as anti‐metastatic agents in the treatment of breast cancer. FEBS letters

550(1-3), 79-83.

Teicher, B. A., Sotomayor, E. A., and Huang, Z. D. (1992). Antiangiogenic agents

potentiate cytotoxic cancer therapies against primary and metastatic disease. Cancer

Research 52(23), 6702-6704.

Page 83: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

68

Tornero-Velez, R., Davis, J., Scollon, E., Starr, J. M., Setzer, R. W., Goldsmith, M.,

Chang, D., Xue, J., Zartarian, V., and DeVito, M. J. (2012). A pharmacokinetic model of

cis-and trans-permethrin disposition in rats and humans with aggregate exposure

application. Toxicological Sciences doi, kfs236.

Tornero-Velez, R., Mirfazaelian, A., Kim, K.-B., Anand, S. S., Kim, H. J., Haines, W. T.,

Bruckner, J. V., and Fisher, J. W. (2010). Evaluation of deltamethrin kinetics and

dosimetry in the maturing rat using a PBPK model. Toxicology and applied

pharmacology 244(2), 208-217.

Tremblay, R. T., Kim, D., and Fisher, J. W. (2012). Determination of tissue to blood

partition coefficients for nonvolatile herbicides, insecticides, and fungicides using

negligible depletion solid-phase microextraction (nd-SPME) and ultrafiltration. Journal of

Toxicology and Environmental Health, Part A 75(5), 288-298.

Willemin, M.-E., Desmots, S., Le Grand, R., Lestremau, F., Zeman, F. A., Leclerc, E.,

Moesch, C., and Brochot, C. (2016). PBPK modeling of the cis-and trans-permethrin

isomers and their major urinary metabolites in rats. Toxicology and applied

pharmacology 294, 65-77.

Wolansky, M., Gennings, C., and Crofton, K. (2006). Relative potencies for acute effects

of pyrethroids on motor function in rats. Toxicological Sciences 89(1), 271-277.

Page 84: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

69

Table 2.1. In Vitro Rat Plasma to RBC Partition Coefficients for DLM (A), CIS (B) and TRANS

(C)

A DLM

Min RBC Plasma PCb

µg/mLa µg/mLa

10 5.9 19.9 3.4

30 6.2 18.2 2.9

45 6.2 18.9 3.0

60 6.0 18.7 3.1

a Mean concentration determined by a modified HPLC technique (n = 2).

b Calculated by dividing the concentration in plasma by concentration in RBC.

B CIS

a Mean concentration determined by a modified HPLC technique (n = 2).

b Calculated by dividing the concentration in plasma by concentration in RBC.

Min RBC Plasma PCb

µg/mLa µg/mLa

10 8.2 11.9 1.5

30 6.8 15.9 2.3

45 6.9 16.0 2.3

60 6.2 18.3 3.0

Page 85: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

70

C TRANS

a Mean concentration determined by a modified HPLC technique (n = 2).

b Calculated by dividing the concentration in plasma by concentration in RBC.

Min RBC Plasma PCb

µg/mLa µg/mLa

10 7.8 12.0 1.5

30 6.5 16.3 2.5

45 6.5 16.2 2.5

60 5.9 18.4 3.1

Page 86: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

71

Table 2.2. DLM (A), CIS (B) and TRANS (C) Concentrations and Tissue:Plasma Partition

Coefficients for Adult Rat Tissues After 72-h Infusiona

A DLM

Tissue DLM (ng/g)b PCc

Plasma 55 ± 17 -

Brain 12 ± 5 0.2 ± 0.1

Liver 27 ± 9 0.6 ± 0.3

Muscle 215 ± 71 3.2 ± 1.1

Fat 3,749 ± 2,357 45 ± 12

a Loading dose = 30 mg/kg po, Infusion rate = 0.36 mg/h for 72 h.

b Tissue concentrations determined by GC-MS are reported as the mean ± SD. (n = 7)

c PC values are reported as the mean ± SD. (n = 7)

B CIS & TRANS

Tissue CIS (ng/g)b PCc TRANS (ng/g)b PCc

Plasma 32 ± 13 - 22 ± 20 -

Brain 21 ± 10 0.6 ± 0.2 14 ± 11 0.7 ± 0.3

Liver 10 ± 6 0.3 ± 0.1 NMd NMd

Muscle 109 ± 83 2.0 ± 0.7 34 ± 9 2.8 ± 1.0

Fat 2,790 ± 877 98 ± 38 1,052 ± 333 67 ± 39

a Loading dose = 150 mg/kg po, Infusion rate = 0.36 mg/h for 72-h.

b Tissue concentrations determined by GC-MS are reported as the mean ± SD. (n = 9)

c PC values are reported as the mean ± SD. (n = 9)

d Not measurable due to low TRANS concentrations.

Page 87: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

72

Table 2.3. Comparison of DLM (A), CIS (B) and TRANS (C) Tissue:Plasma Partition

Coefficients with those Previously Published for Adult Rats.

A DLM

Study Liver Brain Muscle Fat

Current 0.6 0.2 3.2 45

Mirfazaelian et al. 0.4a 0.2 5.6 49

Kim et al. 0.9a 0.2 2.4 46

aIn vivo Tissue:plasma PCs were calculated by dividing the AUC of tissue by AUC of plasma

obtained from a single dose toxicokinetic study.

In Kim et al. (2010) study, plasma and tissue DLM concentration versus time profiles from 0 –

96 h were obtained for single oral dose of 10 mg/kg given to adult SD rats.

In Mirfazaelian et al. (2006) study, plasma and tissue DLM concentration versus time profiles

from 0 – 48 h were obtained for single oral dose of 2 or 10 mg/kg given to adult SD rats.

Page 88: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

73

B CIS

Study Brain Liver Muscle Fat

Current 0.6 0.3 2.0 98

Tornero-Velez et al.

1 mg/kg 5.2a 0.6 NDb 126

Tornero-Velez et al.

10 mg/kg 2.2a 0.5 NDb 87

Willemin et al.

25 mg/kg 2.3a 1.3 1.7 304

aIn vivo Tissue:blood PCs were calculated by dividing the AUC of tissue by AUC of blood

obtained from a single dose toxicokinetic study.

b Not determined in the corresponding study.

In Tornero-Velez et al. (2012) study, plasma and tissue CIS concentration versus time profiles

from 0 – 48 h were obtained for single oral dose of 1 or 10 mg/kg permethrin given to adult Long

Evans rats.

In Willemin et al. (2016) study, plasma and tissue CIS concentration versus time profiles from 0

– 48 h were obtained for single oral dose of 25 mg/kg CIS given to adult SD rats.

Page 89: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

74

C TRANS

Study Brain Liver Muscle Fat

Current 0.7 NMb 2.8 67

Tornero-Velez et al.

1 mg/kg 4.9a 0.5 NDb 58

Tornero-Velez et al.

10 mg/kg 0.6a 0.2 NDc 56

Willemin et al.

25 mg/kg 0.6a NMa 0.77 65

aIn vivo Tissue:blood PCs were calculated by dividing the AUC of tissue by AUC of blood

obtained from a single dose toxicokinetic study.

b Not measurable due to low TRANS concentrations.

c Not determined in the corresponding study.

In Tornero-Velez et al. (2012) study, plasma and tissue TRANS concentration versus time

profiles from 0 – 48 h were obtained for single oral dose of 1 or 10 mg/kg permethrin given to

adult Long Evans rats.

In Willemin et al. (2016) study, plasma and tissue TRANS concentration versus time profiles

from 0 – 48 h were obtained for single oral dose of 25 mg/kg TRANS given to adult SD rats.

Page 90: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

75

Table 2.4. DLM (A), CIS (B), and TRANS (C) Mean Concentrations and Tissue:Plasma

Partition Coefficients for 21-day-old Rats After 48- and 72-h Infusionsa

A DLM

Tissue 48 h 72 h

(ng/g)b PCc (ng/g)d PCe

Plasma 43 ± 28 - 78 ± 47 -

Brain 5 ± 3 0.1 ± 0.01 7 ± 2 0.1 ± 0.03

Liver 4 ± 1 0.1 ± 0.08 6 ± 2 0.1 ± 0.05

Muscle 34 ± 20 0.8 ± 0.2 79 ± 62 1.0 ± 0.7

Fat NDf NDf NDf NDf

a Loading dose = 0.25 mg/kg po, Infusion rate = 0.02 mg/h.

b Tissue levels determined by GC-MS are reported as the mean ± SD. (n = 6)

c PC values are reported as the mean ± SD. (n = 6)

d Tissue levels determined by GC-MS are reported as the mean ± SD. (n = 7)

e PC values are reported as the mean ± SD. (n = 7)

f Not determined due to lack of fat in pups.

No statistically significant differences (p>0.05) in between 48 and 72-h PCs.

Page 91: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

76

B CIS

Tissue 48 h 72 h

(ng/g)b PCc (ng/g)b PCc

Plasma 60 ± 15 - 34 ± 10 -

Brain 38 ± 6 0.7 ± 0.1 34 ± 5 1.0 ± 0.2*

Liver 30 ± 14 0.7 ± 0.3 26 ± 10 0.7 ± 0.3

Muscle 240 ± 73 4.1 ± 1.3 244 ± 83 5.2 ± 1.2

Fat NDd NDd NDd NDd

a Loading dose = 0.25 mg/kg po, Infusion rate = 0.02 mg/h.

b Tissue levels determined by GC-MS are reported as the mean ± SD. (n = 6)

c PC values are reported as the mean ± SD. (n = 6)

d Not determined due to lack of fat in pups.

(*) denotes statistically significant differences (p<0.05) between 48 and 72-h PCs.

Page 92: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

77

C TRANS

Tissue 48 h 72 h

(ng/g)b PCc (ng/g)b PCc

Plasma 58 ± 13 - 28 ± 13 -

Brain 23 ± 4 0.4 ± 0.1 14 ± 6 0.6 ± 0.3

Liver NMd NMd NMd NMd

Muscle 79 ± 27 1.4 ± 0.5 33 ± 12 1.7 ± 0.6

Fat NDe NDe NDe NDe

a Loading dose = 1.15 mg/kg po, Infusion rate = 0.05 mg/h.

b Tissue levels determined by GC-MS are reported as the mean ± SD. (n = 5)

c PC values are reported as the mean ± SD. (n = 5)

d Not measurable due to low TRANS concentrations.

e Not determined due to lack of fat in pups.

No statistically significant differences (p>0.05) in between 48 and 72-h PCs.

Page 93: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

78

Table 2.5. DLM (A), CIS (B), and TRANS (C) Mean Concentrations and Tissue:Plasma

Partition Coefficients for 15-day-old Rats After 48- and 72-h Infusionsa

A DLM

Tissue 48 h 72 h

(ng/g)b PCc (ng/g)b PCc

Plasma 7 ± 5 - 10 ± 9 -

Brain 2 ± 2 0.2 ± 0.09 3 ± 2 0.3 ± 0.1

Liver 9 ± 5 1.1 ± 0.4 12 ± 7 1.6 ± 0.6

Muscle 15 ± 16 2.1 ± 1.1 18 ± 9 1.8 ± 1.0

Fat NDd NDd NDd NDd

a Infusion rate = 0.03 mg/h.

b Tissue levels determined by GC-MS are reported as the mean ± SD. (n = 6)

c PC values are reported as the mean ± SD. (n = 6)

d Not determined due to lack of fat in pups.

No statistically significant differences (p>0.05) in between 48 and 72-h PCs.

Page 94: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

79

B CIS

Tissue 48 h 72 h

(ng/g)b PCc (ng/g)b PCc

Plasma 81 ± 38 - 72 ± 15 -

Brain 76 ± 43 1.1 ± 0.2 82 ± 28 1.1 ± 0.2

Liver 87 ± 46 1.3 ± 0.3 100 ± 13 1.4 ± 0.1

Muscle 194 ± 139 2.5 ± 0.6 290 ± 61 4.1 ± 0.8*

Fat NDd NDd NDd NDd

a Infusion rate = 0.03 mg/h.

b Tissue levels determined by GC-MS are reported as the mean ± SD. (n = 5)

c PC values are reported as the mean ± SD. (n = 5)

d Not determined due to lack of fat in pups.

(*) denotes statistically significant differences (p<0.05) between 48 and 72-h PCs.

Page 95: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

80

C TRANS

Tissue 48 h 72 h

(ng/g)b PCc (ng/g)d PCe

Plasma 80 ± 23 - 52 ± 8 -

Brain 39 ± 6 0.5 ± 0.1 28 ± 5 0.6 ± 0.2

Liver 92 ± 35 1.2 ± 0.4 83 ± 9 1.6 ± 0.4

Muscle 227 ± 104 2.8 ± 1.0 283 ± 33 5.5 ± 1.3*

Fat NDf NDf NDf NDf

a Infusion rate = 0.05 mg/h.

b Tissue levels determined by GC-MS are reported as the mean ± SD. (n = 4)

c PC values are reported as the mean ± SD. (n = 4)

d Tissue levels determined by GC-MS are reported as the mean ± SD. (n = 5)

e PC values are reported as the mean ± SD. (n = 5)

f Not determined due to lack of fat in pups.

(*) denotes statistically significant differences (p<0.05) between 48 and 72-h PCs.

Page 96: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

81

Table 2.6. Comparison of DLM (A), CIS (B), and TRANS (C) Steady-State Tissue:Plasma

Partition Coefficients for Adults, 21-day, and 15-day-old Rats with those Estimated from Single

Dose Toxicokinetic Studies.

A DLM

Tissue 15-day-old 21-day-old Adults

Brain 0.3a

0.4b

0.1

0.1

0.2

0.9

Liver 1.6

1.6

0.1

0.1

0.6

0.8

Muscle 1.8

1.8

1.0

0.8

3.2

0.9

Fat NDc NDc 45

12.8

a In vivo Tissue:plasma PCs at steady-state measured from the current study.

bIn vivo Tissue:plasma PCs calculated by dividing the AUC of tissue by AUC of plasma obtained

from a single dose toxicokinetic study. Plasma and tissue DLM concentration versus time

profiles from 0 – 72 h were obtained for corresponding single oral doses given to PND 15 (0.1

mg/kg), PND 21 (0.1 mg/kg) and adult (0.5 mg/kg) SD rats.

c Not determined due to lack of fat in pups.

Page 97: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

82

B CIS

Tissue 15-day-old 21-day-old Adults

Brain 1.1a

1.0b

1.0

1.3

0.6

1.0

Liver 1.4

2.7

0.7

1.7

0.3

1.6

Muscle 4.1

2.9

5.2

2.1

2.0

2.7

Fat NDc NDc 98

10.6

a In vivo Tissue:plasma PCs at steady-state measured from the current study.

bIn vivo Tissue:plasma PCs were calculated by dividing the AUC of tissue by AUC of plasma

obtained from a single dose toxicokinetic study. Plasma and tissue CIS concentration versus time

profiles from 0 – 24 h were obtained for corresponding single oral doses given to PND 15 (45

mg/kg), PND 21 (45 mg/kg) and adult (120 mg/kg) SD rats.

c Not determined due to lack of fat in pups.

Page 98: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

83

C TRANS

Tissue 15-day-old 21-day-old Adults

Brain 0.6a

0.7b

0.6

0.6

0.7

0.4

Liver 1.6

1.4

NMc

1.1

NMc

0.9

Muscle 5.5

3.6

1.7

2.2

2.8

0.8

Fat NDd NDd 67

4.8

a In vivo Tissue:plasma PCs at steady-state measured from the current study.

bIn vivo Tissue:plasma PCs were calculated by dividing the AUC of tissue by AUC of plasma

obtained from a single dose toxicokinetic study. Plasma and tissue TRANS concentration versus

time profiles from 0 – 24 h were obtained for corresponding single oral doses given to PND 15

(600 mg/kg), PND 21 (600 mg/kg) and adult (300 mg/kg) SD rats.

c Not measurable due to low TRANS concentrations.

d Not determined due to lack of fat in pups.

Page 99: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

84

Figure 2.1. Experimental Approach for In vivo Tissue:Plasma PC Experiment

Page 100: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

85

Brain Liver Muscle0

1

2

3

4

5PND 15

PND 21

Adults

Tis

su

e:p

lasm

a P

C

*#

*#

*

**

Figure 2.2. Comparison of Steady-State Tissue:Plasma PCs for DLM between PND 15,

PND 21 and Adult Rats after 72-h Infusion. (*) denotes statistically significant

differences (p<0.05) compared to adults. (#) denotes statistically significant differences

(p<0.05) compared to PND 21 pups.

Page 101: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

86

Brain Liver Muscle0

1

2

3

4

5

6

7PND 15

PND 21

Adults

Tis

su

e:p

lasm

a P

C

* **#

*

*

*

Figure 2.3. Comparison of Steady-State Tissue:Plasma PCs for CIS between PND 15,

PND 21 and Adult Rats after 72-h Infusion. (*) denotes statistically significant

differences (p<0.05) compared to adults. (#) denotes statistically significant differences

(p<0.05) compared to PND 21 pups.

Page 102: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

87

Brain Liver Muscle0

1

2

3

4

5

6

7PND 15

PND 21

Adults

Tis

su

e:p

lasm

a P

C

*#

*#

Figure 2.4. Comparison of Steady-State Tissue:Plasma PCs for TRANS between PND

15, PND 21 and Adult Rats after 72-h Infusion. (*) denotes statistically significant

differences (p<0.05) compared to adults. (#) denotes statistically significant differences

(p<0.05) compared to PND 21 pups.

Page 103: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

88

CHAPTER 3

EFFECTS OF PLASMA PROTEIN BINDING AND MEMBRANE TRANSPORTERS ON

THE BLOOD BRAIN BARRIER PERMEABILITY OF DELTAMETHRIN2

2Reprinted here with permission from Elsevier. The version of record Manoj A., Anshika

S., Jing P., Srinivasa M., Brian S.C., Catherine A.W., James V.B. and Jason Z. (2016) Toxicology Letters. 250-251; 21-28. is available online at: http://www.sciencedirect.com/science/article/pii/S037842741630042X

Page 104: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

89

ABSTRACT

It is important to assess the extent to which a chemical in the systemic circulation gains

access to the brain, especially for neurotoxic compounds such as pyrethroids. We

evaluated BBB permeability and uptake of DLM into the brain as a function of its free

fraction in plasma, as well as the potential role of membrane transporters. 14C-DLM in

HBSS buffer with 0.01% and 4% human serum albumin (HSA) concentrations was

infused through the left common carotid artery @ 500 µL/min for 2 min, using a Harvard

infusion pump. The left half of the brain was then collected, processed and analyzed for

DLM by liquid scintillation counting. Infusion of DLM in 0.01% and 4% HSA resulted in

left brain levels of 258.3 and 137.0 pmol/g respectively. Cyclosporine A (CSA) was

coinfused with 14C-DLM to examine the potential role of transporters in brain uptake.

CSA reduced the uptake of DLM from 258.3 to 175.1 pmol/g when infused in 0.01%

HSA, whereas in 4% HSA, uptake was not significantly altered. These data suggest that

unidentified influx transporters may play a role in the transport of high concentrations of

free DLM across the BBB. Disruption of BBB by prior infusion of mannitol increased the

uptake of DLM only in the absence of HSA. In summary, these data show the novel

finding that DLM uptake into the brain is dependent on its free fraction in plasma and

the binding to plasma proteins appear to limit the uptake of DLM and play a significant

protective role under normal physiological conditions. The role of transporters in uptake

may only be important at high concentrations of free DLM, which may not be observed

at physiological HSA concentrations.

Page 105: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

90

INTRODUCTION

Pyrethroids are synthetic analogs of pyrethrins, produced from the flowers of

Chrysanthemum cinerariaefolium (Seed, 1973). Pyrethroids are highly lipophilic (Log

Ko/w = 5.7-7.6) compounds with very low water solubility (Laskowski, 2002). The phase

out of organophosphates in the last decade made pyrethroids as the pesticides of

choice in the U.S.A. By 1998, about 25% of the world wide insecticide market was

constituted by pyrethroids (Casida and Quistad, 1998). This dramatic increase in the

use of pyrethroids stems from their relatively low mammalian toxicity and low

environmental persistence. Pyrethroids are often applied to agricultural fields and

commercial nurseries to protect crops, residential structures and lawns to control pests.

Certain pyrethroids are also utilized on livestock to control broad range of ectoparasites

such as mites, headlice and scabies (Anadón et al., 2009; Naeher et al., 2009).

There are several incidents where both occupational and non-occupational

exposures of general populations to pyrethroids have been documented. Non-

occupational exposure occurs predominantly through the ingestion of fruits and

vegetables contaminated with pyrethroids. It was reported that pyrethroid residues were

found in food commodities consumed by elementary school-age children (Lu et al.,

2010). An association was observed between residential environmental levels of

pyrethroids and their metabolite levels in children’s urine. Occupational exposure to

pyrethroids occurs mainly through the dermal route (Bradberry et al., 2005). Increased

levels of 3-phenoxybenzoic acid (3-PBA), a metabolite of pyrethroids, were observed in

Japanese pest control operators (Wang et al., 2007).

Page 106: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

91

Pyrethroids exert neurotoxicity by delaying the closure of voltage-sensitive sodium

channels, thereby prolonging the sodium tail current (Soderlund et al., 2002). The

duration of sodium channel opening and the subsequent clinical manifestations depend

on the structure of pyrethroids. Type I pyrethroids lack the α-cyano group on the alcohol

moiety and their major toxic signs are characterized by tremors and paresthesias (T-

syndrome). Type II pyrethroids possess α-cyano substituent on the phenoxybenzyl

alcohol moiety and include salivation and choreoathetosis (CS-syndrome) as their major

signs of toxicity (Shafer et al., 2005).

The entry of most compounds into the central nervous system (CNS) is impeded by

the presence of blood brain barrier (BBB), which is characterized by the presence of

tight junctions between capillary endothelial cells, lack of fenestrations and limited

pinocytotic transport (Begley and Brightman, 2003). The presence of these tight

junctions close-off the paracellular diffusional pathway and presents a diffusional barrier

for the transport of small, polar solutes. This forces most compounds to cross the BBB

either by lipophilic transcellular diffusion or by specialized carrier mediated transport

mechanisms. Compounds which are small (MW <400), lipophilic, non-polar containing 5

or less hydrogen bond donors or acceptors, and unionized are ideal compounds to

cross the BBB by passive diffusion (Clark, 2003; Van De Waterbeemd et al., 2001).

Deltamethrin (DLM) is a highly lipophilic compound (Log Ko/w = 6.1) and is thus

expected to cross the BBB readily by passive diffusion. However, this might not always

be true due to the binding of these compounds to plasma proteins. It is generally

accepted that plasma protein binding limits the brain uptake and that drug distribution to

the brain is driven by the free, unbound concentration in circulating plasma (Buxton,

Page 107: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

92

2006). Moreover, a large number of membrane transporters expressed at the BBB are

capable of either facilitating or restricting the brain uptake.

To our knowledge, no one has evaluated potential determinants of penetration for

DLM or other pyrethroids across the BBB. There is a lack of understanding of the effect

of plasma protein binding on brain uptake of DLM and the absence of an accurate

quantitative model that appropriately describes this effect. The overall objective of this

study was to understand and evaluate the role of plasma protein binding and membrane

transporters in uptake of DLM into the CNS, so that the exact contribution of these

parameters can be better predicted.

MATERIALS AND METHODS

Materials

Human Serum Albumin (HSA) and mannitol were purchased from Sigma-Aldrich (St.

Louis, MO). Hanks Balanced Salt Solution (HBSS) buffer was purchased from

Mediatech (Manassas, VA). Cyclosporine (CSA) was purchased from Sigma-Aldrich

(Laramie, WY). Dimethyl Sulfoxide (DMSO) was purchased from Fisher chemicals (Fair

Lawn, NJ). Radiolabeled [14C]-DLM (57.9 mCi/mMole) was kindly donated by Bayer

Crop Science (Research Triangle Park, NC).

Animal surgical preparation

Brain uptake of DLM was measured using a modified in situ rat brain perfusion

technique, originally developed by Takasato et al (Boje, 2001; Takasato et al., 1984a).

The University of Georgia Animal Care and Use Committee approved the protocol for

Page 108: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

93

this study. Adult male Sprague-Dawley (SD) rats (300-350g) were purchased from

Charles River Laboratories (Raleigh, NC). Upon receipt, all animals were inspected by a

qualified animal technician. The rats were quarantined and their health monitored for

one week at the University of Georgia’s AAALAC-accredited central animal facility. Each

rat was housed in a cage with a 12-h light/dark cycle at ambient temperature (22ºC) and

relative humidity (55 ± 5%). Food (5001 Rodent Diet, PMI Nutrition LLC, Brentwood,

MO) and water were provided ad libitum. Rats were anesthetized with KAX (Ketamine,

Acepromazine, and Xylazine) cocktail. The left common carotid artery was exposed and

cannulated with a 25G needle affixed to PE-50 tubing for perfusion fluid administration.

Prior to the start of the infusion, the heart was stopped by rapidly severing the cardiac

ventricles to eliminate contributions of vascular perfusion from the systemic circulation.

The left common carotid artery was then perfused with 2 mL of sterile saline to flush

blood from the brain. This was followed by the infusion of corresponding perfusion

fluids.

Processing of brain

Since the perfusion fluid was infused into the left common carotid artery, only the left

cerebral hemisphere was processed for DLM quantitation. The isolated brain tissue was

homogenized with a Tekmar Tissumizer IKA Ultra-Turrax homogenizer (Janke and

Kunkel Laboratories, Cridersville, OH), by adding two volumes of ice-cold distilled water

by weight. An aliquot (100 µL) of the brain homogenate was added to 4 mL of EcoLite

scintillation cocktail (MP Biomedicals, Solon, OH). Uptake of DLM was quantified by

measuring the radioactivity in each sample by liquid scintillation counting using a

Beckman Coulter LS 6500 (Brea, CA) and normalizing to tissue weight.

Page 109: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

94

Effect of HSA and CSA on DLM associated brain levels

The perfusion fluid consisted of 10 μM [14C]-DLM and 0.01 or 4% HSA with or without

the P-gp inhibitor CSA (25 μM) in HBSS buffer. All solutions were incubated for 15 min

at 37°C prior to the infusion. In the CSA group, animals were pre-treated with CSA prior

to the start of DLM infusion by perfusing 2 mL of sterile saline containing 25 μM CSA.

The DLM containing perfusion fluid (including 25 μM CSA) was then infused at a

constant rate of 500 μL/min for 2 min using a Harvard 22 syringe infusion pump

(Harvard Apparatus, Holliston, MA).

Role of the BBB in limiting DLM associated brain levels

Mannitol (1.6 M) was infused at 0.25 mL/kg/sec for 30 seconds prior to the perfusion of

DLM to disrupt the BBB (Brown et al., 2004). The perfusion fluid consisting of 1 μM

[14C]-DLM with or without 4% HSA in HBSS buffer was then infused at a constant rate of

500 μL/min for 2 min using a Harvard 22 syringe infusion pump.

Statistical Analysis

All experiments were performed with a minimum of three independent experiments

unless otherwise stated. Statistical significance was evaluated between groups using

either the Student’ t-test or one way ANOVA, followed by Tukey’s multiple comparison

test, as indicated, with a significance level of p<0.05, using Graphpad Prism 6 software.

Page 110: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

95

RESULTS

Effect of HSA and CSA on DLM associated brain levels

To assess the influence of plasma protein binding and to evaluate the relative

contribution of carrier-mediated transport to BBB permeability of DLM, the brain uptake

was measured from 0.01% and 4% HSA in the presence and absence of CSA (Fig 3.1).

The brain uptake of DLM, when infused from 0.01% and 4% HSA was 258.3 pmol/g and

137.0 pmol/g, respectively. The uptake from 4% HSA was significantly lower as

compared to 0.01% HSA (p<0.05). Interestingly, prior infusion of CSA reduced the

uptake only from 0.01% HSA (P<0.05), whereas from 4% HSA, the uptake was not

significantly altered (p>0.05).

Role of the BBB in limiting DLM associated brain levels

To understand the role of BBB and plasma protein binding in limiting the entry of DLM

into the CNS, the uptake of DLM was measured with and without 4% HSA, in control

and mannitol treated animals (Fig 3.2). As compared to control animals, prior treatment

with mannitol significantly increased the uptake (p<0.05) from HBSS buffer, whereas

from 4% HSA, the uptake was not altered significantly (p>0.05).

DISCUSSION

Compounds with high lipophilicity are long known to penetrate the BBB easily. Thus,

DLM, which is a highly lipophilic compound (Log Ko/w = 6.1) is expected to penetrate the

BBB with relative ease. However, DLM peak brain concentrations and AUCs were found

to be only 20% of plasma values (Kim et al., 2008). The measured brain:plasma PC for

Page 111: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

96

DLM at steady-state was only 0.2 and quite low (Amaraneni et al., to be submitted). It

was reported that ideally, a compound should have a Log Ko/w value in the range of 1-3,

to have good brain permeation (Dishino et al., 1983; Van De Waterbeemd et al., 2001).

DLM has a molecular weight (MW) of 505.2 and a Log Ko/w value of 6.1, which might

have satisfied the criteria for poor brain permeation. Apart from these two criteria,

plasma protein binding also plays a major role in restricting the brain uptake of DLM. It

was reported that DLM is about 90% bound to adult human plasma (Sethi et al., 2014).

Only about 10% of DLM is free and available for diffusion across the BBB.

Conventionally, only unbound or free fraction of a compound is available for uptake

into tissues (Pardridge, 2001). Pardridge et al. reported that the brain extraction of

propranolol, measured using the brain uptake index technique, decreased when the

albumin concentrations are increased (Pardridge and Landaw, 1984). Marathe et al.

examined the effect of serum protein binding on the distribution of brain lidocaine in

dogs. A strong and positive correlation was observed between brain to serum and

serum free fractions (Marathe et al., 1991). Rowley et al. reported that the binding of the

glycine/NMDA receptor antagonists to plasma protein limits their brain penetration

(Rowley et al., 1997). Several other studies have reported good agreement between

unbound fraction in the plasma and brain concentrations (Cory Kalvass and Maurer,

2002; Dubey et al., 1989).

An in situ brain perfusion technique was used in these studies to measure the

brain DLM uptake. This technique was originally developed by Takasato et al (Takasato

et al., 1984b). As opposed to brain uptake index (BUI) technique, perfusion time can be

varied (10 – 600 sec) much longer than that of a single pass (1 – 2 sec), allowing the

Page 112: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

97

BBB permeability to be measured over an extended range. The contributions from

peripheral metabolism are eliminated by severing the heart and by pre-perfusing for 30-

90 sec to wash out vascular constituents. Finally, not only is this technique sensitive,

but also allows for absolute control over the perfusate composition, and thus, allows us

to quantify the effect of different protein concentrations and inhibitors on brain uptake by

varying the concentrations in the perfusate (Murakami et al., 2000). In the present

study, the brain uptake of DLM from 0.01% HSA was significantly lower than from 4%

HSA. These results suggest that brain influx for DLM correlated well with the free

fraction in the plasma. These results are consistent with results from in vitro studies

performed by Dr. Zastre (see Appendix Fig 2). It was demonstrated that accumulation

into human cerebral microvessel endothelial cells (hCMEC/D3) increased with

increasing DLM unbound fraction. We also conducted an experiment, in which a

hypertonic solution of mannitol was infused into the carotid artery of anesthetized rats in

order to assess the influence of physical disruption of the BBB on the subsequent brain

uptake of DLM. Mannitol infusion produced about 2.2 fold increase in the brain uptake

of DLM in the absence of 4% HSA. Virtually, 100% of the DLM is present as the free or

unbound fraction under these conditions. Brain uptake of DLM was not significantly

altered by mannitol pre-treatment when it was infused with 4% HSA. Only 20% of DLM

is present as unbound fraction in the presence of 4% HSA. Albumin, of course, is too

large a molecule (MW ̴66,000) to cross the BBB, even when the tight junctions between

the endothelial cells are disrupted by hypertonic mannitol (Brown et al., 2004). The

observation that mannitol failed to significantly enhance brain uptake of DLM in the

presence of 4% HSA, even when the BBB integrity is compromised, illustrates that

Page 113: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

98

plasma protein binding limits uptake of this neuroactive insecticide, and plays a

significant protective role under normal physiological conditions.

In addition to the diffusion barrier, the BBB also presents a transport barrier in

the form of multiple carrier-mediated influx and efflux transporters at the brain capillary

apical and basolateral membranes. While p-glycoprotein (p-gp) is the major efflux

transporter located at the luminal membrane, organic anion transporting polypeptides

(OATPs) and organic anion transporters (OATs) are the major solute carriers located at

both the luminal and abluminal membranes (Abbott et al., 2006; Lee et al., 2001). It

was reported that the uptake of DLM into caco-2 cells was not restricted by p-gp efflux

but undergoes absorptive influx transport (Zastre et al., 2013). However, nothing is

known about the role of p-gp or other influx transporters at the BBB. Interestingly, prior

infusion of CSA significantly reduced the DLM uptake from 0.01% HSA, but not from 4%

HSA. At 0.01% HSA, a higher free fraction of DLM is available to interact with the

transporters, whereas at 4% HSA, the free fractions are not high enough for CSA to

significantly impact the uptake. The fact that the DLM uptake was reduced but not

increased by CSA casts doubt on the contributions of some unidentified influx

transporters. However, specific transporter family or isoform involved in the transport

could not be identified from these studies and requires further research. It should be

remembered that despite being a major p-gp inhibitor, CSA is also known to be a

promiscuous inhibitor of both influx and efflux transporters (Letschert et al., 2006). Our

results are well supported by results from in vitro studies (Appendix). DLM uptake into

hCMEC/D3 cells from 0.01% HSA was inhibited in the presence of CSA, which is an

indication of a specific influx transport process.

Page 114: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

99

In vitro results also demonstrate that substantial differences in the DLM

accumulation into hCMEC/D3 cells can only be observed up to 1% HSA concentration.

No significant difference was observed in the uptake between 2% and 4% HSA. Human

albumin concentrations range from as low as 2.9% in neonates to as high as 4% in

adults. From in vitro results, although it appears that differences in brain uptake of DLM

would not be anticipated between human neonates and adults based on differences in

albumin concentrations, confirmatory evidence from future in situ studies would be

needed in this regard. Regardless of these findings, there is no data available on the

effect of lipoprotein binding on the brain uptake of DLM, which is an important player in

real life scenario. This may have additional implications related to the significance and

physiological utility of these studies. Thus, it would be interesting to see how the binding

of DLM to both albumin and lipoproteins together in the rat and human plasma, and how

the age-related changes in these protein concentrations affect brain uptake. Apart from

the ontogeny of plasma proteins, ontogeny of BBB penetration can also influence the

brain influx. Therefore, the future studies will determine the brain uptake of DLM as a

function of age-dependent changes in plasma protein binding and BBB penetration.

In conclusion, our results indicate that BBB plays some role in limiting the entry of

DLM into the brain. Apart from BBB, plasma protein binding is also instrumental in

restricting the entry of DLM into brain under normal physiological conditions. Thus,

extensive binding to physiological albumin concentrations resulting in lower free

fractions, is the primary reason why brain levels of DLM were lower than anticipated.

Also, we found that p-gp does not play a role in limiting the DLM uptake into the brain. A

low affinity influx transporter is suggested to contribute to DLM uptake, which becomes

Page 115: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

100

significant only at high free fractions not typically observed under normal physiological

conditions. The DLM influx into the brain is a function of its free fraction in the plasma.

Page 116: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

101

REFERENCES

Abbott, N. J., Rönnbäck, L., and Hansson, E. (2006). Astrocyte–endothelial interactions

at the blood–brain barrier. Nature Reviews Neuroscience 7(1), 41-53.

Anadón, A., Martínez-Larrañaga, M., and Martínez, M. (2009). Use and abuse of

pyrethrins and synthetic pyrethroids in veterinary medicine. The Veterinary Journal

182(1), 7-20.

Begley, D. J., and Brightman, M. W. (2003). Structural and functional aspects of the

blood-brain barrier. In Peptide transport and delivery into the central nervous system

(doi, pp. 39-78. Springer.

Boje, K. M. (2001). In vivo measurement of blood-brain barrier permeability. Curr Protoc

Neurosci Chapter 7, Unit 7 19, 10.1002/0471142301.ns0719s15.

Bradberry, S. M., Cage, S. A., Proudfoot, A. T., and Vale, J. A. (2005). Poisoning due to

pyrethroids. Toxicological reviews 24(2), 93-106.

Brown, R. C., Egleton, R. D., and Davis, T. P. (2004). Mannitol opening of the blood–

brain barrier: regional variation in the permeability of sucrose, but not 86 Rb+ or

albumin. Brain research 1014(1), 221-227.

Page 117: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

102

Buxton, I. L. (2006). Pharmacokinetics and pharmacodynamics: the dynamics of drug

absorption, distribution, action, and elimination. Goodman & Gilman's: the

pharmacological basis of therapeutics. 11th ed. New York: McGraw-Hill 11.

Casida, J. E., and Quistad, G. B. (1998). Golden age of insecticide research: past,

present, or future? Annual review of entomology 43(1), 1-16.

Clark, D. E. (2003). In silico prediction of blood–brain barrier permeation. Drug

discovery today 8(20), 927-933.

Cory Kalvass, J., and Maurer, T. S. (2002). Influence of nonspecific brain and plasma

binding on CNS exposure: implications for rational drug discovery. Biopharmaceutics &

drug disposition 23(8), 327-338.

Dishino, D., Welch, M. J., Kilbourn, M. R., and Raichle, M. E. (1983). Relationship

between lipophilicity and brain extraction of C-11-labeled radiopharmaceuticals. Journal

of nuclear medicine: official publication, Society of Nuclear Medicine 24(11), 1030-1038.

Dubey, R., McAllister, C., Inoue, M., and Wilkinson, G. (1989). Plasma binding and

transport of diazepam across the blood-brain barrier. No evidence for in vivo enhanced

dissociation. Journal of Clinical Investigation 84(4), 1155.

Page 118: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

103

Kim, K.-B., Anand, S. S., Kim, H. J., White, C. A., and Bruckner, J. V. (2008).

Toxicokinetics and tissue distribution of deltamethrin in adult Sprague–Dawley rats.

Toxicological sciences 101(2), 197-205.

Laskowski, D. A. (2002). Physical and chemical properties of pyrethroids. In Reviews of

environmental contamination and toxicology (doi, pp. 49-170. Springer.

Lee, G., Dallas, S., Hong, M., and Bendayan, R. (2001). Drug transporters in the central

nervous system: brain barriers and brain parenchyma considerations. Pharmacological

reviews 53(4), 569-596.

Letschert, K., Faulstich, H., Keller, D., and Keppler, D. (2006). Molecular

characterization and inhibition of amanitin uptake into human hepatocytes. Toxicological

sciences 91(1), 140-149.

Lu, C., Schenck, F. J., Pearson, M. A., and Wong, J. W. (2010). Assessing children's

dietary pesticide exposure: direct measurement of pesticide residues in 24-hr duplicate

food samples. Environmental health perspectives 118(1), 1625-1630.

Marathe, P. H., Shen, D. D., Artru, A. A., and Bowdle, T. A. (1991). Effect of serum

protein binding on the entry of lidocaine into brain and cerebrospinal fluid in dogs.

Anesthesiology 75(5), 804-812.

Page 119: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

104

Murakami, H., Takanaga, H., Matsuo, H., Ohtani, H., and Sawada, Y. (2000).

Comparison of blood-brain barrier permeability in mice and rats using in situ brain

perfusion technique. American Journal of Physiology-Heart and Circulatory Physiology

279(3), H1022-H1028.

Naeher, L. P., Barr, D. B., Rithmire, N., Edwards, J., Holmes, A. K., Needham, L. L.,

and Rubin, C. S. (2009). Pesticide exposure resulting from treatment of lice infestation

in school-aged children in Georgia. Environment international 35(2), 358-362.

Pardridge, W. M. (2001). Brain drug targeting: the future of brain drug development.

Cambridge University Press.

Pardridge, W. M., and Landaw, E. M. (1984). Tracer kinetic model of blood-brain barrier

transport of plasma protein-bound ligands. Empiric testing of the free hormone

hypothesis. Journal of Clinical Investigation 74(3), 745.

Rowley, M., Kulagowski, J. J., Watt, A. P., Rathbone, D., Stevenson, G. I., Carling, R.

W., Baker, R., Marshall, G. R., Kemp, J. A., and Foster, A. C. (1997). Effect of plasma

protein binding on in vivo activity and brain penetration of glycine/NMDA receptor

antagonists. Journal of medicinal chemistry 40(25), 4053-4068.

Seed, P. (1973). Pyrethrum: the natural insecticide doi.

Page 120: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

105

Sethi, P. K., Muralidhara, S., Bruckner, J. V., and White, C. A. (2014). Measurement of

plasma protein and lipoprotein binding of pyrethroids. Journal of pharmacological and

toxicological methods 70(1), 106-111.

Shafer, T. J., Meyer, D. A., and Crofton, K. M. (2005). Developmental neurotoxicity of

pyrethroid insecticides: critical review and future research needs. Environmental health

perspectives 113(2), 123.

Soderlund, D. M., Clark, J. M., Sheets, L. P., Mullin, L. S., Piccirillo, V. J., Sargent, D.,

Stevens, J. T., and Weiner, M. L. (2002). Mechanisms of pyrethroid neurotoxicity:

implications for cumulative risk assessment. Toxicology 171(1), 3-59.

Takasato, Y., Rapoport, S. I., and Smith, Q. R. (1984a). An in situ brain perfusion

technique to study cerebrovascular transport in the rat. American Journal of Physiology

247(3, Pt. 2), H484-H493.

Takasato, Y., Rapoport, S. I., and Smith, Q. R. (1984b). An in situ brain perfusion

technique to study cerebrovascular transport in the rat. Am J Physiol 247(3 Pt 2), H484-

H493.

Van De Waterbeemd, H., Smith, D. A., Beaumont, K., and Walker, D. K. (2001).

Property-based design: optimization of drug absorption and pharmacokinetics. Journal

of medicinal chemistry 44(9), 1313-1333.

Page 121: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

106

Wang, D., Kamijima, M., Imai, R., Suzuki, T., Kameda, Y., Asai, K., Okamura, A., Naito,

H., Ueyama, J., and Saito, I. (2007). Biological monitoring of pyrethroid exposure of pest

control workers in Japan. Journal of occupational health 49(6), 509-514.

Zastre, J., Dowd, C., Bruckner, J., and Popovici, A. (2013). Lack of P-glycoprotein-

mediated efflux and the potential involvement of an influx transport process contributing

to the intestinal uptake of deltamethrin, cis-permethrin, and trans-permethrin.

toxicological sciences doi, kft193.

Page 122: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

107

0.01% 0.01% + CSA 4% 4% + CSA0

100

200

300

400p

mo

l/g

bra

in

*

*

Figure 3.1. Effect of HSA and CSA on the In vivo Brain Uptake of DLM. The Left

common carotid artery of adult male Sprague-Dawley rats was cannulated and

infused (500 µL/min) with 10 µM 14C-DLM in 0.01% and 4% HSA solutions either

alone or in combination with 25 µM CSA for 2 min. Results represent the mean ± SD

(n = 4 - 7). (*) denotes statistically significant differences (p<0.05).

Page 123: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

108

HBSS 4% HSA0

25

50

75

100

125

Control Mannitol

pm

ol/

g b

rain

**

*

Figure 3.2. Effect of Mannitol on the In vivo Brain Association of DLM With and

Without 4% HSA. The Left common carotid artery of adult male Sprague-Dawley

rats was cannulated and infused (500 μL/min) with 1 μM 14C-DLM with and without

4% HSA. In the mannitol group, animals were pre-treated with 1.6 M Mannitol prior

to infusion of DLM. Results represent the mean ± SD (n = 4 - 5). (*, and **) denotes

statistically significant differences (p<0.05).

Page 124: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

109

CHAPTER 4

BRAIN UPTAKE OF DELTAMETHRIN IN RATS AS A FUNCTION OF AGE

DEPENDENT CHANGES IN PLASMA PROTEIN BINDING AND BBB

PENETRATION3

3Manoj A., Jing P., Srinivasa M., Tanzir B.M., Brian S.C., Catherine A.W., and James V.B. To be submitted to Drug Metabolism and Disposition.

Page 125: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

110

ABSTRACT

For potentially neurotoxic compounds such as pyrethroids, it is important to assess the

extent to which a chemical in the systemic circulation gains access to the brain. This

study investigated the role of age-dependent differences in plasma protein binding and

BBB permeability on brain uptake of DLM using an in situ brain perfusion technique. We

evaluated uptake of DLM into the brain from different physiological concentrations of

HSA (2 – 5%). No significant differences were observed in the uptake of DLM among

the groups tested. We measured uptake from plasma of PND 15, PND 21 and adult

rats, and found no significant difference. Uptake of DLM from human plasma from birth

– 1 week, 1 – 4 weeks, 4 weeks – 1 year, 1 – 3 years and adults was not significantly

different among each group. We assessed the influence of individual lipoprotein

constituents on the brain entry of DLM by measuring its uptake from physiological

concentrations of LDL (100 mg/dL), HDL (50 mg/dL), and their mixture. The % DLM

bound was similar among these groups and as was the uptake. We measured uptake

from different LDL concentrations (50 – 400 mg/dL) to understand the influence of

changes in LDL concentrations on brain uptake in hyper-cholesterolemia patients. The

uptake into the brain was reduced with increasing LDL concentrations, although it was

statistically significant only at 400 mg/dL. We also assessed brain uptake of DLM as a

function of the maturation of the BBB. Uptake of three different concentrations of DLM

from 4% HSA was measured in PND 15, PND 21 and adult rats. At all three

concentrations tested, uptake in PND 15 and PND 21 pups was significantly greater

than in adults. While PND 15 pups exhibited about a 2 – 4 fold increase in uptake

compared to PND 21 and adult rats, PND 21 pups exhibited about a 2 fold increase in

Page 126: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

111

uptake relative to adults. This data demonstrate that the brain uptake of environmental

concentrations of DLM is not influenced by age dependent differences in plasma protein

binding. However, these studies revealed that, at physiological albumin concentrations,

uptake of DLM into the brain is a function of maturation of the BBB and is inversely

proportional to age. The younger the rat, the greater the uptake. This increased uptake

in younger rats may result from increased permeability of the BBB in this age group.

Page 127: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

112

INTRODUCTION

Pyrethroids are the synthetic derivatives of pyrethrins obtained from the flowers

of Chrysanthemum cinerariaefolium. Combined with the reduced use of

organophosphates and their relatively lower mammalian toxicity, the use of pyrethroids

has dramatically increased over the last decade (Power and Sudakin, 2007). The uses

of pyrethroids are many fold and are often used as insecticides in agricultural,

commercial and residential settings. Medically, they are used in the treatment of

scabies, mites and headlice on pets and humans (Naeher et al., 2009). Several

occupational and non-occupational exposures of large segments of the populace have

been documented, although at low levels. The most frequent route of exposure is

ingestion of pyrethroids from dust and through eating foods containing residues of these

chemicals (Baker et al., 2000). About 75% of the urine samples sampled from a large

German population were found to have pyrethroid metabolites (Heudorf et al., 2004).

Flight attendants working on commercial flights disinfected with pyrethroids were found

to have elevated urinary levels of pyrethroid metabolites compared to those that did not

use pyrethroids (Wei et al., 2012). Low levels of these chemicals may be found in fruits

and vegetables because of their application to crops (Lu et al., 2010).

Pyrethroids are classified as type I and type II pyrethroids based on their

chemical structure and toxic signs from acute poisoning. Type II pyrethroids have an α-

cyano substituent on the 3-phenoxybenzyl alcohol moiety. Pyrethroids primarily act on

voltage-sensitive sodium channels in the central nervous system and exert neurotoxicity

by delaying the closure of sodium channel gates (Soderlund et al., 2002). Type I

pyrethroids delay the closure of sodium channels gates only for few milliseconds

Page 128: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

113

resulting in repetitive nerve discharges, whereas the type II pyrethroids delay it for

several seconds resulting in stimulus-dependent nerve depolarization and blockage

(Shafer et al., 2005). This difference in the duration of modification results in

characteristic clinical manifestations of type I and type II pyrethroids poisoning. Toxic

signs of type I pyrethroid poisoning are characterized by hyperexcitation, fine tremors

followed by whole body tremors and paresthesia, typically named as “T-syndrome”.

Type II pyrethroids are characterized by excessive salivation, coarse tremors

progressing to choreoathetosis and clonic seizures, collectively referred to as “CS-

syndrome” (Ray, 2001).

The BBB characterized by extensive tight junctions and the absence of

fenestrations, acts as a barrier which restricts the entry of most compounds from blood

to brain (Ballabh et al., 2004). In general, compounds with high lipophilicity, small

molecular size and low plasma protein binding diffuse freely across the BBB (Clark,

2003). Despite being highly lipophilic (Log P = 6.1), the entry of DLM into the brain is

limited by its high molecular weight (505.2) and high plasma protein binding (80%). Only

unbound compound is free to diffuse across the BBB. The brain uptake of DLM is

influenced by their extent of plasma protein binding and is reduced with a decrease in

its free fraction (fu) (Amaraneni et al., 2016). Plasma proteins show age-dependent

differences between neonates and adults, in terms of their concentration and binding

affinity (Alcorn and McNamara, 2003). Neonates have lower plasma protein binding

compared to adults resulting in higher free fractions. It is important to assess the

influence of age-dependent differences in the plasma protein binding on the brain

uptake of DLM. By examining the effect of binding to various concentrations of albumin

Page 129: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

114

and lipoproteins in the physiological range, it is possible to understand how the brain

uptake of DLM varies in individuals with disease conditions such as hypoalbuminemia

or hypercholesterolemia.

Immature rats were more sensitive to the potential neurotoxicity of high doses of

pyrethroids (Sheets et al., 1994). This can be attributed, in part, to the immaturity of the

BBB along with other immature physiological and biochemical processes that alter the

brain dosimetry. In general, the BBB in neonates is immature, poorly formed and leaky.

The BBB undergoes several structural changes during development before becoming

fully functional. Thus, the development of the BBB is a gradual process in humans,

which is more immature and permeable at birth and becomes fully functional by

approximately 6 months of age (Watson et al., 2006).

There is very little information available in the literature on the differences in the

BBB permeability of pyrethroids between younger and adult age-groups. No studies

have been conducted regarding how the binding of DLM to albumin and individual

lipoprotein constituents such as LDL and HDL, affect its brain permeability. The effect of

age-dependent differences in such plasma proteins and BBB penetration has also never

been investigated. The objective of the current project was to examine the influence of

age-dependent differences in the plasma protein binding and the BBB penetration on

the brain uptake of DLM using an in situ brain perfusion technique. The effect of binding

to albumin and lipoproteins constituents on the brain uptake was investigated. Such a

determination of BBB permeability can be incorporated into physiologically based

pharmacokinetic (PBPK) models for prediction of brain dosimetry in humans of different

ages.

Page 130: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

115

MATERIALS AND METHODS

Materials

Human Serum Albumin (HSA) was purchased from Golden West Biologicals

(Temecula, CA). Low-density lipoproteins (LDL) and high-density lipoproteins (HDL)

were purchased from Lee Biosolutions, Inc. (Maryland Heights, MO). Rat plasma of

different age groups was purchased from Innovative Research, Inc. (Novi, MI). Hanks

Balanced Salt Solution (HBSS) buffer was purchased from Mediatech (Manassas, VA).

Radiolabeled [14C]-DLM (57.9 mCi/mMole) was kindly donated by Bayer Crop Science

(Research Triangle Park, NC). EcoLite scintillation cocktail was provided by MP

Biochemicals (Solon, OH). Acetonitrile (HPLC-grade), hexamethyldisilazane (Reagent-

grade), sodium fluoride (NaF) (purity, 99.0 %) were purchased from Sigma-Aldrich (St.

Louis, MO). Isooctane (purity, 99.0 %) and 2-Octanol (Laboratory-grade) were

purchased from Fisher Scientific (Pittsburgh, PA).

Human Plasma Samples

Anonymized plasma samples were obtained from Cincinnati Children’s Hospital

Medical Center (CCHMC) Biobank in Cincinnati, OH. Refrigerated plasma samples that

were not used for clinical studies were pooled, frozen at -20°C, and accumulated for

each of the following age-groups: 0 – 1 week; 1 – 4 weeks; 4 weeks – 1 year; 1 – 3

years. Adult plasma was purchased from Innovative Research, Inc. (Novi, MI).

Specimens from pediatric donors suffering from blood diseases were excluded. The

health of the subjects was not otherwise verified. Gender and ethnicity were not

Page 131: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

116

inclusion criteria. The use of the anonymous plasma samples for the current research

project was reviewed and approved by the CCHMC Tissue Use Committee.

In vitro binding studies of DLM

Binding of DLM was determined by a three step solvent extraction method

developed by Sethi et al. (Sethi et al., 2014). Binding to different HSA concentrations (2

– 5%), physiological lipoprotein concentrations (100 mg/dL LDL, 50 mg/dL HDL, and

their mixture), and different LDL concentrations (50, 200, and 400 mg/dL) was

determined to correlate % free DLM to brain uptake. Briefly, a volume of 90 µL of

corresponding biological matrix (HSA or lipoproteins) was spiked with 10 µL of 10 µM

[14C]-DLM in silanized glass vials. Samples were then incubated in an orbital shaker at

37°C with constant shaking (100 rpm) for 15 min for HSA and 30 min for lipoproteins.

After 15 or 30 min, samples were removed and 200 µL of isooctane was added to each

tube to extract the free (unbound) compound. The samples were vortexed for 30 sec

and centrifuged at 10,000 rpm for 10 min. Three 50 µL aliquots were removed of the

isooctane layer and transferred to scintillation vials containing 4 mL of EcoLiteTM

scintillation cocktail and vortexed for 10 sec. After removing the remaining isooctane,

300 µL of acetonitrile was added to each tube to extract the total bound compound. The

tubes were then vortexed and centrifuged as mentioned above. A 50 µL aliquot was

taken in triplicates and transferred to a scintillation vial with EcoLiteTM and vortexed. The

tubes were counted for radioactivity by LS 6500 Liquid Scintillation counter.

The in situ brain perfusion solutions were incubated for 15 – 30 min prior to

infusion. To determine if this short time of incubation affects the binding of DLM. The

Page 132: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

117

different biological matrices (human plasma, HSA, and lipoproteins) used in perfusion

studies, were spiked with [14C]-DLM and incubated for 15, 30, 60, 120, 180 and 240 min

before the extraction. After the appropriate time of incubation, the above extraction

procedure was repeated and counted for radioactivity. Since human plasma contains

both albumin and lipoproteins, after performing isooctane extraction and before

proceeding to acetonitrile extraction, tubes were extracted with n-octanol to determine

the fraction bound to lipoproteins. After isooctane extraction was done, 200 µL of n-

octanol was added to each tube, vortexed and centrifuged as above. Three 50 µL

aliquots were then removed and added to the EcoLite cocktail in scintillation vials,

vortex and counted for radioactivity.

Animal surgical preparation

Brain uptake of DLM was measured using a modified in situ rat brain perfusion

technique, originally developed by Takasato et al (Boje, 2001; Takasato et al., 1984).

The University of Georgia Animal Care and Use Committee approved the protocol for

this study. Adult male Sprague-Dawley (SD) rats (300-350 g) and pregnant female SD

rats were purchased from Charles River Laboratories (Raleigh, NC). Upon receipt, all

animals were inspected by a qualified animal technician. The rats were quarantined and

their health monitored for one week at the University of Georgia’s AAALAC-accredited

central animal facility. For immature BBB studies, the pups were allowed to reach 15 or

21 days of age before being used for experiments. Each rat was housed in a cage with

a 12-h light/dark cycle at ambient temperature (22ºC) and relative humidity (55 ± 5%).

Food (5001 Rodent Diet, PMI Nutrition LLC, Brentwood, MO) and water were provided

ad libitum. Rats were anesthetized with KAX (Ketamine, Acepromazine, and Xylazine)

Page 133: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

118

cocktail. The left common carotid artery was exposed and cannulated with a 23G

needle affixed to PE-50 tubing for perfusion fluid administration. In experiments with

pups, due to the small diameter of the carotid artery, a 27G needle affixed to PE-10

tubing was used for perfusion fluid administration.

Perfusion protocol

Prior to the start of the infusion, the cardiac ventricles were rapidly severed to ensure

the elimination of flow contributions from the systemic circulation. The left common

carotid artery was then perfused with 2 mL and 0.5 mL of sterile saline in adults and

pups respectively, to remove blood. This was followed by the infusion of the

corresponding perfusion fluids. Once the infusion was stopped, the left common carotid

artery was again perfused with corresponding volumes of sterile saline to remove

unabsorbed DLM in the vasculature.

Processing of brain

Since the perfusion fluid was infused into the left common carotid artery, only the left

cerebral hemisphere was processed for DLM quantitation. The isolated brain tissue was

homogenized with a Tekmar Tissumizer IKA Ultra-Turrax homogenizer (Janke and

Kunkel Laboratories, Cridersville, OH), by adding two volumes of ice-cold distilled water

by weight. An aliquot (1000 µL) of the brain homogenate was added to 4 mL of EcoLite

scintillation cocktail (MP Biomedicals, Solon, OH). Uptake of DLM was quantified by

measuring the radioactivity in each sample by liquid scintillation counting using a

Beckman Coulter LS 6500 (Brea, CA) and normalizing to tissue weight.

Page 134: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

119

Effect of binding to physiological HSA

Uptake of DLM into brain from different physiological HSA concentrations was

measured in adult SD rats. The perfusion fluid consisted of 1 µM [14C]-DLM in 2%,

2.9%, 4% or 5% HSA solutions made in HBSS buffer. All solutions were incubated for

15 min at 37°C in an orbital shaker, prior to the infusion. The perfusion fluid was then

infused at a constant rate of 500 µL/min for 2 minutes using a Harvard 22 syringe

infusion pump (Harvard Apparatus, Holliston, MA).

Effect of binding to physiological LDL and HDL

The brain uptake of DLM from physiological concentrations of LDL (100 mg/dL), HDL

(50 mg/dL) and their mixture (100 mg/dL LDL and 50 mg/dL HDL) was measured from

adult SD rats. A control group was also included, where the uptake of DLM was

measured from HBSS buffer alone. Brain uptake was also measured from the LDL

concentrations of 50, 200, and 400 mg/dL to mimic uptake in individuals with

hypocholesterolemia and hypercholesterolemia. The perfusion fluid consisted of 1 µM

[14C]-DLM in corresponding lipoprotein solutions made in HBSS buffer. All solutions

were incubated for 30 min at 37°C in an orbital shaker prior to the infusion. The

perfusion fluid was then infused at a constant rate of 500 µL/min for 2 minutes using a

Harvard 22 syringe infusion pump (Harvard Apparatus, Holliston, MA).

Effect of age-dependent differences in plasma protein binding

The uptake of DLM from rat plasma and human plasma of different age groups was

measured in adult SD rats. For uptake studies from rat plasma, the perfusion fluid

consisted of 1 µM [14C]-DLM in PND 15, PND 21 or adult rat plasma. For uptake studies

Page 135: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

120

from human plasma, the perfusion fluid consisted of 1 µM [14C]-DLM in human plasma

of ages birth - 1 week, 1 - 4 weeks, 4 weeks - 1 year, 1 – 3 years, and adults. All

solutions were incubated for 15 min at 37°C in an orbital shaker, prior to the infusion.

The perfusion fluid was then infused at a constant rate of 500 µL/min for 2 minutes

using a Harvard 22 syringe infusion pump (Harvard Apparatus, Holliston, MA).

Effect of age-dependent differences in BBB penetration

The uptake of different concentrations of DLM was measured in PND 15, PND 21 and

adult rats. The perfusion fluid consisted of either 1, 10 or 50 µM [14C]-DLM in 4% HSA

solution made in HBSS buffer. All solutions were incubated for 15 min at 37°C in an

orbital shaker, prior to the infusion. The perfusion fluid was then infused at a constant

rate of 500 µL/min for 2 minutes using a Harvard 22 syringe infusion pump (Harvard

Apparatus, Holliston, MA). The flow rate was reduced to 250 µL/min and the infusion

time was increased to 4 minutes in PND 15 and PND 21 pups to make sure that the

perfusion pressure in microvessels was not too high and the integrity of BBB was not

compromised. A control study was conducted in adult rats to make sure that the change

in flow rate did not alter the uptake of DLM. As part of the control study, 1 µM [14C]-DLM

from 4% HSA was infused in adult SD rats at a flow rate of 250 µL/min for 4 minutes.

Data analysis

All experiments were performed with a minimum of three independent animals unless

otherwise stated. Statistical significance was evaluated between groups using either

student’s t-test or one way ANOVA, followed by Dunnett’s or Tukey’s multiple

Page 136: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

121

comparison test, as indicated, with a significance level of p<0.05, using Graphpad Prism

5 software.

RESULTS

Effect of incubation time on the binding of DLM

These in vitro experiments were conducted to determine the influence of incubation

time on the binding of DLM to different biological matrices that were used in animal

perfusion studies. No significant change in binding of DLM to human plasma was

observed across the incubation times tested (Fig 4.1). The same pattern was observed

in the binding of DLM to 4% HSA, except that the binding increased to 71% after 240

min of incubation (Fig 4.2). However, there was an initial increase in LDL binding from

48% to 63% after 30 min of incubation (Fig 4.3). No further changes were observed

after 30 min. There were no significant time related changes in the binding of DLM to

HDL (Fig 4.4) and the lipoprotein mixture (Fig 4.5). The percent bound remained the

same throughout the incubation periods.

Impact of binding to physiological albumin concentrations

In order to assess the influence of physiological changes in albumin concentration on

the brain uptake of DLM, experiments were conducted to measure uptake from 2 - 5%

HSA. Brain uptake (pmol/g) of DLM from these experiments is shown in Fig 4.6.

Interestingly, the uptake of DLM did not vary significantly between 2% and 5% HSA

(p>0.05). This finding was supported by the fact that there was no significant difference

in the binding of DLM between 2 – 5% HSA (Fig 4.7). The bound DLM increased from

Page 137: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

122

41% in 2% HSA to 49% in 5% HSA. This increase was not significant enough to affect

the uptake into the brain.

Effect of binding to physiological lipoprotein concentrations

To establish the effect of binding to individual lipoprotein constituents i.e. LDL and

HDL on the BBB permeability of DLM, the uptake of DLM into the brain from

physiological concentrations of LDL (100 mg/dL), HDL (50 mg/dL) and their mixture was

measured (Fig 4.8). Even though the uptake from LDL and the mixture was slightly

lower than that of HDL, the difference was not significant (p>0.05). The uptake from

LDL, HDL, and the mixture was slightly lower as compared to HBSS buffer, but again

not significantly different (p>0.05). It can be observed in Fig 4.9 that there was no

significant difference in the binding of DLM to LDL (48%), HDL (51%), and the mixture

(58%) (p>0.05). Thus, there was not much change in free fraction between these

groups to affect the brain uptake.

To mimic the uptake in individuals with hypocholesterolemia and

hypercholesterolemia, the brain uptake of DLM from 50, 200, and 400 mg/dL LDL was

measured (Fig 4.10). The uptake of DLM decreased with increased LDL concentration.

The lowest uptake was measured from 400 mg/dL, which was significantly different from

that of 50 and 100 mg/dL (p<0.05). Even though the uptake from 200 mg/dL was lower

than 50 and 100 mg/dL, the difference failed to reach statistical significance and so was

between 50 and 100 mg/dL. In Fig 4.11, it can be seen that the binding of DLM was

highest at 400 mg/dL (83%) and was significantly different from that of 50 (61%), 100

(55%) and 200 mg/dL (66%) concentrations.

Page 138: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

123

Impact of age-dependent differences in plasma protein binding

To determine the influence of age-dependent differences in plasma protein binding

on the brain uptake of DLM, the uptake was measured from human and rat plasma of

different age groups. In Fig 4.12, it can be seen that the uptake of DLM from 15-day rat

plasma (9.1 pmol/g) was not significantly different to that of 21-day (12.5 pmol/g) and

adult rat plasma (9.7 pmol/g) (p>0.05). Sethi et al. (Sethi et al., 2015) reported that the

total binding of DLM to adult (80%) and 21-day-old rats (79%) was only slightly higher

than that of 15-day-old rats (73%).

Similar to rat plasma, the uptake from human plasma did not change much from

birth - 1 week (13.6 pmol/g) to adults (13.5 pmol/g) (Fig 4.13). Thus, the difference in

uptake from human plasma was not significantly different between the age-groups

tested (p>0.05). Sethi et al. (Sethi et al., 2015) reported that the total binding of DLM to

human plasma of ages birth – 1 week (73%) and 1 – 4 weeks (77%) was significantly

lower compared to adults (90%). However, binding did not change significantly in other

age groups. Interestingly, such differences were not observed in our in situ uptake

studies.

Impact of age-dependent differences in BBB penetration

To examine how the differences in BBB penetration affect the brain entry of DLM,

the uptake of different concentrations of DLM was measured by cannulating PND 15,

PND 21, and adult SD rats (Fig 4.14). The uptake of DLM was significantly higher in

PND 15 rats compared to PND 21 and adult rats at all concentrations tested (p<0.05).

Similarly, PND 21 rats exhibited significantly greater DLM uptake compared to adults at

Page 139: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

124

10 and 50 µM (p<0.05). Although this was also true at 1 µM DLM, the difference failed

to reach statistical significance. The magnitude of difference in the uptake between PND

15 and adult rats was greatest at 1 µM concentration, while for 21-day rats, it remained

constant at all concentrations tested (Table 4.1). A control study was conducted in adult

rats to see if the change in flow rate affects the uptake into the brain. In Fig 4.16, it can

be observed that the uptake was slightly decreased from 12.8 pmol/g to 9.3 pmol/g

when the flow rate was reduced from 500 µL/min to 250 µL/min. However, this decrease

was not statistically significant (p>0.05).

The brain uptake of increasing concentrations of DLM from 4% HSA in all the

three age-groups is shown in Fig 4.15. These results suggest that the uptake of DLM

from 4% HSA was linear at the concentration range tested in all the three age groups.

DISCUSSION

There are primarily three mechanisms by which a compound can enter the brain.

They are passive diffusion, carrier-mediated (facilitative), and ATP-dependent (active)

transport processes (Lee et al., 2001). The experiments from our previous work have

indicated that DLM is not a substrate of p-glycoprotein (p-gp). An uncharacterized low

affinity influx transporter is suggested to contribute to DLM uptake by the brain, which is

significant only at higher free fractions of DLM, which is not typically observed either in

adults or younger age groups (Amaraneni et al., 2016). Thus, the ontogenic differences

in the transporters do not contribute to the age-dependent differences in the brain

dosimetry of DLM. Therefore, two factors, namely lower plasma protein binding and an

Page 140: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

125

immature BBB, are considered to be important determinants of greater transport of DLM

across the BBB in younger age-groups.

The free fraction of a compound in plasma is an important determinant of the extent

to which a compound diffuses freely across the BBB along its concentration gradient.

Our previous work established that the in situ brain uptake of DLM from 0.01% HSA

was significantly greater than that of 4% HSA, confirming that brain uptake is dependent

on the free fraction of DLM. Interestingly, the uptake of DLM into human cerebral

microvessel endothelial cells (hCMEC/D3) did not vary significantly between 2% - 4%

HSA (Amaraneni et al., 2016). The HSA concentrations in humans range from 4% in

adults and to 2.9% in neonates at birth (Sethi et al., 2015). Therefore, we chose to

measure the in situ brain uptake of DLM from 2% - 5% HSA to see how the binding of

DLM to the physiological range of HSA affects its brain uptake. This would tell us not

only about the effect of age-dependent differences in HSA concentration on the brain

uptake of DLM, but also how it varies in individuals with hypo- or hyper-albuminemia.

Similar to our previous in vitro findings, the uptake of DLM into brain did not vary

significantly between 2% and 5% HSA. The bound DLM at 2% HSA (41%) was not

significantly different from that of 5% HSA (49%). These results suggest that the age-

dependent differences in HSA concentration do not significantly affect the brain uptake

of low levels of DLM. Either extremely low levels of HSA (<2%) or very high

concentrations of DLM are required to impact the brain uptake significantly, both of

which are rare to occur physiologically.

Since DLM is a highly lipophilic compound and exhibits a high degree of binding to

lipoproteins in plasma, we focused on how the binding of DLM to individual lipoprotein

Page 141: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

126

constituents such as LDL, HDL and their mixture affect its brain uptake. No studies have

been undertaken to examine the influence of binding to individual lipoprotein

constituents on the brain uptake of pyrethroids. These experiments were conducted at a

normal physiological range of LDL and HDL. The brain uptake of DLM from

physiological concentrations of LDL, HDL, and their mixture did not vary significantly

from each other. Although the uptake from the lipoprotein mixture (50 mg/dL HDL and

100 mg/dL) was lower than the uptake from HDL (50 mg/dL), it was not statistically

significant. It should be noted that the corresponding free fractions of DLM did not vary

significantly between these groups. Unexpectedly, despite the same free fractions, the

DLM uptake from physiological concentrations of lipoproteins was much higher

compared to the uptake from physiological HSA concentrations. This might be attributed

to the presence of lipoprotein receptors on the surface of brain capillary endothelial cells

(ECs). It is also possible that DLM has a higher binding affinity for albumin than

lipoprotein constituents due to simple partitioning into hydrophobic regions of

lipoproteins. There is growing evidence that the BBB is equipped with LDL receptors for

maintaining lipid and cholesterol homeostasis (Méresse et al., 1989). Pitas et al. (Pitas

et al., 1987) demonstrated the presence of apoB,E (LDL) receptors in rat and monkey

brains using immunocytochemistry. They also reported that apolipoprotein E and apo A-

I were present in the human cerebrospinal fluid (CSF). LDL receptor mediated

transcytosis is the primary mechanism for lipid transport and cholesterol homeostasis in

the central nervous system. Dehouck et al. (Dehouck et al., 1997) used a coculture

model of bovine brain capillary endothelial cells and rat astrocytes to demonstrate the

transport of 125I-LDL and acetylated 125I-LDL through brain capillary EC monolayers.

Page 142: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

127

They reported that LDL was transcytosed through the BBB via a receptor mediated

mechanism. This mechanism is successfully employed in drug delivery to the brain

using nanoparticles (Kreuter, 2001). Nanoparticles coated with polysorbates serve as

an anchor for apo E and adsorb them onto their surface. These nanoparticles with apo

E on their surface mimic LDL particles and interact with LDL receptors leading to their

uptake by the ECs.

There has been a dramatic increase in hypercholesterolemic adults in the United

States (Ford et al., 2010). Consequently, the use of cholesterol lowering drugs such as

statins has also increased rapidly (Kit, 2014). Since hypercholesterolemia is usually

associated with increased LDL levels, we examined the brain uptake of DLM from

higher LDL concentrations to get a glimpse in patients with hypercholesterolemia. The

uptake into the brain decreased with increasing LDL concentrations with the lowest

uptake measured at 400 mg/dL. This can be correlated to the lowest free fraction of

DLM at 400 mg/dL, which was almost half when compared to the other concentrations

tested. These results corroborate our previous finding that the uptake of DLM into the

brain is a function of its free fraction. However, caution must be employed when

extrapolating these findings to human hypercholesterolemic patients. Since under

normal physiological conditions, these elevated levels of LDL always coexist with

albumin and other lipoproteins, it is difficult to assess the cumulative effect of binding to

all of these plasma proteins on the brain uptake. At least, these results suggest that the

brain uptake of DLM may be slightly lower in individuals with hypercholesterolemia as

compared to those with normal LDL levels.

Page 143: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

128

Plasma proteins show age-dependent differences in terms of their concentration

and binding affinity. Neonatal animals have relatively low plasma protein binding

capacity due to low albumin levels, lower binding affinity, and elevated levels of bilirubin,

fatty acids and other substances that compete with xenobiotics for binding sites

(Ginsberg et al., 2004). Plasma albumin levels in neonates are about 75% of adult

human levels, while α1 acid-glycoprotein concentrations are 75% of those in adults by 6

months (Alcorn and McNamara, 2003). Marked age-dependent differences in plasma

binding may result in a significant effect on the pyrethroids’ BBB permeability and their

brain dosimetry. In contrast, neonatal rats have about 50% of the adult plasma proteins,

albumin and α1 acid-globulins (de Zwart et al., 2008). Thus, the plasma protein binding

capacity of neonatal and preweanling rats for pyrethroids is likely significantly lower than

for their human counterparts. Therefore, we have chosen to measure the brain uptake

of DLM from human and rat plasma of different age groups separately.

Sethi et al. (Sethi et al., 2015) reported that the total plasma binding of DLM was

about 80% in both 21-day-old and adult rats, while it was approximately 75% in 15-day-

old rats. Even though the difference in binding between 15-day-old and adult rats was

reported to be statistically significant, the difference in the free fraction was not high

enough to impact the brain uptake. We did not observe any significant difference in the

in situ brain uptake of DLM from the plasma of 15-day, 21-day, and adult rats. Sethi et

al. (Sethi et al., 2015) reported that significant differences in the binding of DLM to

human plasma were observed only up to 4 weeks of age, after which the binding was

reported to be similar to that of adults. The binding of DLM in human plasma of ages 1

week and 4 weeks was reported to be approximately 75%, which was lower compared

Page 144: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

129

to adults (90%). We measured the uptake of DLM from human plasma of ages birth – 1

week, 1 – 4 weeks, 4 weeks – 1 year, 1 – 3 years, and adults. Interestingly, our results

indicated that there was no significant difference in the brain uptake of DLM between

any of the age groups tested. Together, these results suggest that the age-dependent

differences in plasma protein binding do not significantly impact the brain uptake of DLM

at low exposure levels.

There are only limited data on the stages of human development at which the

permeability of the maturing BBB becomes comparable to that of adults. Some

investigators conclude that recent morphological, biochemical and molecular data

provide substantial evidence for the existence of well developed, effective barrier

mechanisms in the fetus and newborn (Ek et al., 2012). Saunders et al. (Saunders et

al., 2012) reported that intercellular tight junctions between cerebral endothelial cells are

functionally effective as soon as they differentiate. These investigators do note that

some barrier mechanisms may work to a lesser extent in maturing humans and

laboratory animals, though other processes exhibit a higher rate of function.

Vascularization of the brain and BBB maturation have been studied in detail in the

rat. Several researchers conducted early ultrastructural studies of embryonic and

postnatal morphological changes of the cerebral microvasculature of rats (Bär and

Wolff, 1972; Caley and Maxwell, 1970; Schulze and Firth, 1992). The investigators

monitored maturation of the primary structural components of the BBB, including the

vessel wall endothelium, basement membrane, pericytes and astrocytes. Structural

integrity and maturity were reported by each research group to be achieved by PND 21.

The magnitude of microvascular sprouting largely paralleled an increase in the cerebral

Page 145: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

130

cortical mass during these 21 days (Donahue and Pappas, 1961; Rowan and Maxwell,

1981). Stewart and Hayakawa (Stewart and Hayakawa, 1987) described a progressive

reduction in the number and width of clefts between adjacent cerebral capillary

endothelial cells, which paralleled a decrease in the BBB permeability to horseradish

peroxidase in maturing mice. The developmental tightening of the endothelial junctions

was associated with an increasing thickness of the basement membrane and

envelopment by pericytes. Basement membrane, pericytes and astrocytes signaling

induce and maintain the integrity of the endothelial tight junctions (Liebner et al., 2011).

Thus, these cells’ structural and functional development must occur in concert with that

of the endothelium. Ferguson and Woodbury (Ferguson and Woodbury, 1969)

monitored cerebral uptake of inulin in rats as an index of BBB penetration. Brain uptake

of the water soluble compound progressively decreased with increasing age from PND

4-26. Leibnar et al. (Liebner et al., 2011) emphasized the BBB tightness is not “switched

on” at a particular point during brain angiogenesis, but occurs as a gradual process

during embryogenesis and postnatal maturation.

The immature BBB in neonatal rats resulted in higher levels of glucocorticoids

compared to adults (Arya et al., 2006). Upon injection of trypan blue, mice of 4 weeks of

age incorporated more dye compared to mice of 5-8 weeks of age (Ribatti et al., 2006).

Kyu-bong Kim et al. reported that brain DLM concentration versus time profiles are age-

dependent and that the youngest rats exhibited the highest brain concentrations over

time, upon ingestion of DLM (Kim et al., 2010). The BBB was not fully developed

structurally and was not fully functional until about the time of weanling. Interendothelial

junctions in the immature BBB are characterized by membrane separations greater than

Page 146: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

131

20 nm and are leaky (Schulze and Firth, 1992). These leaky interendothelial junctions in

developing brain results in high permeability. The maturation of the BBB is

accompanied by the disappearance of these membrane separations and establishment

of complex tight junctions. Thus, the structural changes at tight junctional contacts

underlie the developmental tightening of the BBB. We measured the brain uptake of

DLM in 15-day, 21-day and adult rats by cannulating their carotid artery.

We have controlled for the free fraction and measured the uptake of three

different concentrations of DLM by cannulating PND 15, PND 21 and adult rats. At all

the concentrations tested, the youngest rats exhibited the highest uptake of DLM.

Interestingly, the uptake of 1 µM DLM in 15-day-old rats was 3.7 fold higher than that of

adults, while for 10 and 50 µM DLM, it was 2.2 and 2.5 fold higher respectively.

Similarly, the magnitude of difference in the uptake between PND 15 and PND 21 rats

was 2.1 and highest at the lowest DLM concentration, while it remained constant at

about 1.5 fold difference at the other two DLM concentrations. It was possible that the

uncharacterized influx transporter at the BBB was saturated at higher DLM

concentrations and thus, had a constant contribution to the DLM uptake. The uptake in

PND 21 rats is about 1.5 fold higher than that of adults at all the concentrations tested.

These results suggest that, the younger the rats, the greater is the uptake. The linear

uptake of DLM in the concentration range tested in all the three age groups indicates

that passive diffusion is the main transport process involved.

Several studies in pediatric patients involving intravenous drug injection and CSF

monitoring have failed to provide definitive information on the age-dependence of

permeability changes of the BBB or brain-CSF barriers (Heideman et al., 1989; Kellie et

Page 147: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

132

al., 2002; Lowe et al., 2001). Jacobs et al. (Jacobs et al., 1986) administered Imipenem

and Cilastatin by i.v. infusion and assessed CSF drug concentrations. No correlation

was seen between age and the extent of CSF penetration. In an early study, Widell

(Widell, 1958) measured concentrations of several proteins in the CSF of 98 normal

children aged 0 – 13 years. Total protein levels diminished during the first 9 months of

life. Similar findings were subsequently reported by other investigators (Statz and

Felgenhauer, 1983; Wong et al., 2000). Both groups of clinicians found that CSF

protein concentrations decreased rapidly during the initial 6 months postnatally. Shah et

al. (Shah et al., 2011) measured CSF protein levels in a large group (n = 375) of infants

only 56 days of age and younger. Levels were quite variable but lowest during the first 2

- 4 weeks after birth. An inverse linear relationship was reported between entry of

sodium fluorescein into the CSF and age of subjects up to 6 months old (Misra et al.,

1987). The largest drop in CSF fluorescein levels occurred during initial 2 weeks of life.

Increased BBB permeability to unconjugated bilirubin is an important contributor to

neonatal jaundice (Brito et al., 2014). Higher brain bilirubin levels were found in PND 2

than in PND 14 piglets given the compound intravenously (Lee et al., 1995). In

summary, functional maturation of the BBB as a barrier to substances in the blood

appears to occur earlier in humans than rats. If it is assumed that maximal barrier

function is largely achieved in both rats and humans within 3 to 4 weeks of parturition,

this interval represents a considerably larger proportion of the rat’s period of

development.

In conclusion, our results indicate that the brain uptake of DLM is a function of age

and is significantly higher in younger age groups. The more immature the animal, the

Page 148: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

133

less efficient is the BBB in limiting the uptake of DLM into the brain. At low exposure

levels of DLM, the ontogeny of plasma proteins does not significantly affect its brain

uptake. Likewise, changes in physiological concentrations of albumin or lipoproteins

would not have any significant impact on the brain uptake of low levels of DLM. Extreme

hypoalbuminemic conditions or very high concentrations of DLM would be needed to

have a significant effect on the brain uptake.

Acknowledgement: The authors would like to thank the Council for the Advancement

of Pyrethroid Human Risk Assessment (CAPHRA) for funding of this work. MA was

partially supported by a University of Georgia Interdisciplinary Toxicology Program

graduate stipend.

Page 149: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

134

REFERENCES

Alcorn, J., and McNamara, P. J. (2003). Pharmacokinetics in the newborn. Advanced

drug delivery reviews 55(5), 667-686.

Amaraneni, M., Sharma, A., Pang, J., Muralidhara, S., Cummings, B. S., White, C. A.,

Bruckner, J. V., and Zastre, J. (2016). Plasma protein binding limits the blood brain

barrier permeation of the pyrethroid insecticide, deltamethrin. Toxicology letters 250, 21-

28.

Arya, V., Demarco, V. G., Issar, M., and Hochhaus, G. (2006). Contrary to adult,

neonatal rats show pronounced brain uptake of corticosteroids. Drug metabolism and

disposition 34(6), 939-942.

Baker, S. E., Barr, D. B., Driskell, W. J., D BEESON, M., and Needham, L. L. (2000).

Quantification of selected pesticide metabolites in human urine using isotope dilution

high-performance liquid chromatography/tandem mass spectrometry. Journal of

Exposure Science and Environmental Epidemiology 10, 789-798.

Ballabh, P., Braun, A., and Nedergaard, M. (2004). The blood–brain barrier: an

overview: structure, regulation, and clinical implications. Neurobiology of disease 16(1),

1-13.

Page 150: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

135

Bär, T., and Wolff, J. (1972). The formation of capillary basement membranes during

internal vascularization of the rat's cerebral cortex. Zeitschrift für Zellforschung und

Mikroskopische Anatomie 133(2), 231-248.

Boje, K. M. (2001). In vivo measurement of blood-brain barrier permeability. Curr Protoc

Neurosci Chapter 7, Unit 7 19, 10.1002/0471142301.ns0719s15.

Brito, M. A., Palmela, I., Cardoso, F. L., Sá-Pereira, I., and Brites, D. (2014). Blood–

Brain Barrier and Bilirubin: Clinical Aspects and Experimental Data. Archives of medical

research 45(8), 660-676.

Caley, D. W., and Maxwell, D. S. (1970). Development of the blood vessels and

extracellular spaces during postnatal maturation of rat cerebral cortex. Journal of

Comparative Neurology 138(1), 31-47.

Clark, D. E. (2003). In silico prediction of blood–brain barrier permeation. Drug

discovery today 8(20), 927-933.

de Zwart, L., Scholten, M., Monbaliu, J. G., Annaert, P. P., Van Houdt, J. M., Van den

Wyngaert, I., De Schaepdrijver, L. M., Bailey, G. P., Coogan, T. P., and Coussement,

W. C. (2008). The ontogeny of drug metabolizing enzymes and transporters in the rat.

Reproductive Toxicology 26(3), 220-230.

Page 151: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

136

Dehouck, B., Fenart, L., Dehouck, M.-P., Pierce, A., Torpier, G., and Cecchelli, R.

(1997). A new function for the LDL receptor: transcytosis of LDL across the blood–brain

barrier. The Journal of cell biology 138(4), 877-889.

Donahue, S., and Pappas, G. D. (1961). The fine structure of capillaries in the cerebral

cortex of the rat at various stages of development. American Journal of Anatomy

108(3), 331-347.

Ek, C. J., Dziegielewska, K. M., Habgood, M. D., and Saunders, N. R. (2012). Barriers

in the developing brain and Neurotoxicology. Neurotoxicology 33(3), 586-604.

Ferguson, R. K., and Woodbury, D. M. (1969). Penetration of 14C-inulin and 14C-

sucrose into brain, cerebrospinal fluid, and skeletal muscle of developing rats.

Experimental brain research 7(3), 181-194.

Ford, E. S., Li, C., Pearson, W. S., Zhao, G., and Mokdad, A. H. (2010). Trends in

hypercholesterolemia, treatment and control among United States adults. International

journal of cardiology 140(2), 226-235.

Ginsberg, G., Hattis, D., and Sonawane, B. (2004). Incorporating pharmacokinetic

differences between children and adults in assessing children's risks to environmental

toxicants. Toxicology and applied pharmacology 198(2), 164-183.

Page 152: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

137

Heideman, R. L., Cole, D. E., Balis, F., Sato, J., Reaman, G. H., Packer, R. J., Singher,

L. J., Ettinger, L. J., Gillespie, A., and Sam, J. (1989). Phase I and pharmacokinetic

evaluation of thiotepa in the cerebrospinal fluid and plasma of pediatric patients:

evidence for dose-dependent plasma clearance of thiotepa. Cancer research 49(3),

736-741.

Heudorf, U., Angerer, J., and Drexler, H. (2004). Current internal exposure to pesticides

in children and adolescents in Germany: urinary levels of metabolites of pyrethroid and

organophosphorus insecticides. International archives of occupational and

environmental health 77(1), 67-72.

Jacobs, R. F., Kearns, G., Brown, A., and Longee, D. (1986). Cerebrospinal fluid

penetration of imipenem and cilastatin (primaxin) in children with central nervous

system infections. Antimicrobial agents and chemotherapy 29(4), 670-674.

Kellie, S. J., Barbaric, D., Koopmans, P., Earl, J., Carr, D. J., and de Graaf, S. S.

(2002). Cerebrospinal fluid concentrations of vincristine after bolus intravenous dosing.

Cancer 94(6), 1815-1820.

Kim, K.-B., Anand, S. S., Kim, H. J., White, C. A., Fisher, J. W., Tornero-Velez, R., and

Bruckner, J. V. (2010). Age-, Dose-and Time-Dependency of Plasma and Tissue

Distribution of Deltamethrin in Immature Rats. Toxicological Sciences doi, kfq074.

Page 153: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

138

Kit, B. K. (2014). Prescription cholesterol-lowering medication use in adults aged 40 and

over: United States, 2003–2012 doi.

Kreuter, J. (2001). Nanoparticulate systems for brain delivery of drugs. Advanced drug

delivery reviews 47(1), 65-81.

Lee, C., Stonestreet, B. S., Oh, W., Outerbridge, E. W., and Cashore, W. J. (1995).

Postnatal maturation of the blood-brain barrier for unbound bilirubin in newborn piglets.

Brain research 689(2), 233-238.

Lee, G., Dallas, S., Hong, M., and Bendayan, R. (2001). Drug transporters in the central

nervous system: brain barriers and brain parenchyma considerations. Pharmacological

reviews 53(4), 569-596.

Liebner, S., Czupalla, C. J., and Wolburg, H. (2011). Current concepts of blood-brain

barrier development. International Journal of Developmental Biology 55(4-5), 467-476.

Lowe, E. S., Kitchen, B. J., Erdmann, G., Stork, L. C., Bostrom, B. C., Hutchinson, R.,

Holcenberg, J., Reaman, G. H., Woods, W., and Franklin, J. (2001). Plasma

pharmacokinetics and cerebrospinal uid penetration of thioguanine in children with

acute lymphoblastic leukemia: a collaborative Pediatric Oncology Branch, NCI, and

Children's Cancer Group study. Cancer Chemother Pharmacol 47, 199-205.

Page 154: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

139

Lu, C., Schenck, F. J., Pearson, M. A., and Wong, J. W. (2010). Assessing children's

dietary pesticide exposure: direct measurement of pesticide residues in 24-hr duplicate

food samples. Environmental health perspectives 118(1), 1625-1630.

Méresse, S., Delbart, C., Fruchart, J. C., and Cecchelli, R. (1989). Low‐density

lipoprotein receptor on endothelium of brain capillaries. Journal of neurochemistry 53(2),

340-345.

Misra, P., Gulati, A., Mahesh, A., Sharma, B., Malik, G., and Dhawan, K. (1987).

Maturity of blood brain barrier in children. The Indian journal of medical research 85,

401.

Naeher, L. P., Barr, D. B., Rithmire, N., Edwards, J., Holmes, A. K., Needham, L. L.,

and Rubin, C. S. (2009). Pesticide exposure resulting from treatment of lice infestation

in school-aged children in Georgia. Environment international 35(2), 358-362.

Pitas, R., Boyles, J., Lee, S., Hui, D., and Weisgraber, K. (1987). Lipoproteins and their

receptors in the central nervous system. Characterization of the lipoproteins in

cerebrospinal fluid and identification of apolipoprotein B, E (LDL) receptors in the brain.

Journal of Biological Chemistry 262(29), 14352-14360.

Page 155: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

140

Power, L. E., and Sudakin, D. L. (2007). Pyrethrin and pyrethroid exposures in the

United States: a longitudinal analysis of incidents reported to poison centers. Journal of

medical toxicology 3(3), 94-99.

Ray, D. (2001). Pyrethroid insecticides: mechanisms of toxicity, systemic poisoning

syndromes, paresthesia, and therapy. Handbook of pesticide toxicology 2, 1289-1303.

Ribatti, D., Nico, B., Crivellato, E., and Artico, M. (2006). Development of the blood‐

brain barrier: A historical point of view. The Anatomical Record Part B: The new

Anatomist 289(1), 3-8.

Rowan, R. A., and Maxwell, D. S. (1981). Patterns of vascular sprouting in the postnatal

development of the cerebral cortex of the rat. American Journal of Anatomy 160(3),

247-255.

Saunders, N. R., Liddelow, S. A., and Dziegielewska, K. M. (2012). Barrier mechanisms

in the developing brain. Frontiers in pharmacology 3, 46.

Schulze, C., and Firth, J. A. (1992). Interendothelial junctions during blood-brain barrier

development in the rat: morphological changes at the level of individual tight junctional

contacts. Developmental brain research 69(1), 85-95.

Page 156: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

141

Sethi, P. K., Muralidhara, S., Bruckner, J. V., and White, C. A. (2014). Measurement of

plasma protein and lipoprotein binding of pyrethroids. Journal of pharmacological and

toxicological methods 70(1), 106-111.

Sethi, P. K., White, C. A., Cummings, B. S., Hines, R. N., Muralidhara, S., and

Bruckner, J. V. (2015). Ontogeny of Plasma Proteins, Albumin and Binding of

Diazepam, Cyclosporine and Deltamethrin. Pediatric research doi.

Shafer, T. J., Meyer, D. A., and Crofton, K. M. (2005). Developmental neurotoxicity of

pyrethroid insecticides: critical review and future research needs. Environmental health

perspectives 113(2), 123.

Shah, S. S., Ebberson, J., Kestenbaum, L. A., Hodinka, R. L., and Zorc, J. J. (2011).

Age‐specific reference values for cerebrospinal fluid protein concentration in neonates

and young infants. Journal of hospital medicine 6(1), 22-27.

Sheets, L. P., Doherty, J. D., Law, M. W., Reiter, L. W., and Crofton, K. M. (1994). Age-

dependent differences in the susceptibility of rats to deltamethrin. Toxicology and

applied pharmacology 126(1), 186-190.

Soderlund, D. M., Clark, J. M., Sheets, L. P., Mullin, L. S., Piccirillo, V. J., Sargent, D.,

Stevens, J. T., and Weiner, M. L. (2002). Mechanisms of pyrethroid neurotoxicity:

implications for cumulative risk assessment. Toxicology 171(1), 3-59.

Page 157: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

142

Statz, A., and Felgenhauer, K. (1983). Development of the Blood‐CSF Barrier.

Developmental Medicine & Child Neurology 25(2), 152-161.

Stewart, P., and Hayakawa, E. (1987). Interendothelial junctional changes underlie the

developmental ‘tightening’of the blood-brain barrier. Developmental Brain Research

32(2), 271-281.

Takasato, Y., Rapoport, S. I., and Smith, Q. R. (1984). An in situ brain perfusion

technique to study cerebrovascular transport in the rat. American Journal of Physiology

247(3, Pt. 2), H484-H493.

Watson, R. E., DeSesso, J. M., Hurtt, M. E., and Cappon, G. D. (2006). Postnatal

growth and morphological development of the brain: a species comparison. Birth

Defects Research Part B: Developmental and Reproductive Toxicology 77(5), 471-484.

Wei, B., Mohan, K. R., and Weisel, C. P. (2012). Exposure of flight attendants to

pyrethroid insecticides on commercial flights: urinary metabolite levels and implications.

International journal of hygiene and environmental health 215(4), 465-473.

Widell, S. (1958). On the Cerebrospinal Fluid in Normal Children and in Patients with

Acute Abacterial Meningo‐Encephalitis. Acta Paediatrica 47(6), 711-713.

Page 158: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

143

Wong, M., Schlaggar, B. L., Buller, R. S., Storch, G. A., and Landt, M. (2000).

Cerebrospinal fluid protein concentration in pediatric patients: defining clinically relevant

reference values. Archives of pediatrics & adolescent medicine 154(8), 827-831.

Page 159: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

144

15 30 60 120 2400

10

20

30

40

50

60

70

80

90

Incubation Time (Min)

% B

ou

nd

*

Figure 4.1. Effect of Time of Incubation on In vitro Binding of DLM to Human Plasma.

One µM 14

C-DLM in adult human plasma was incubated for 15, 30, 60, 120, and 240

min at 37°C in an orbital shaker, before extracting with isooctane and acetonitrile.

Results represent the Mean ± SD (n = 3). (*) represents statistically significant

difference (p<0.05) compared to 15 min.

Page 160: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

145

15 30 60 120 2400

10

20

30

40

50

60

70

80

Incubation Time (Min)

% B

ou

nd

Figure 4.2. Effect of Time of Incubation on In vitro Binding of DLM to HSA. One µM 14

C-

DLM in 4% HSA was incubated for 15, 30, 60, 120, and 240 min at 37°C in an orbital

shaker, before extracting with isooctane and acetonitrile. Results represent the Mean ±

SD (n = 3). There was no statistically significant difference in the % bound DLM

compared to 15 min (p>0.05).

Page 161: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

146

15 30 60 120 2400

10

20

30

40

50

60

70

80

Incubation Time (Min)

% B

ou

nd

Figure 4.3. Effect of Time of Incubation on In vitro Binding of DLM to LDL. One µM 14

C-

DLM in 100 mg/dL LDL was incubated for 15, 30, 60, 120, and 240 min at 37°C in an

orbital shaker, before extracting with isooctane and acetonitrile. Results represent the

Mean ± SD (n = 3). There was no statistically significant difference in the % bound DLM

compared to 15 min (p>0.05).

Page 162: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

147

15 30 60 120 2400

10

20

30

40

50

60

Incubation Time (Min)

% B

ou

nd

Figure 4.4. Effect of Time of Incubation on In vitro Binding of DLM to HDL. One µM 14

C-

DLM in 50 mg/dL HDL was incubated for 15, 30, 60, 120, and 240 min at 37°C in an

orbital shaker, before extracting with isooctane and acetonitrile. Results represent the

Mean ± SD (n = 3). There was no statistically significant difference in the % bound DLM

compared to 15 min (p>0.05).

Page 163: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

148

15 30 60 120 2400

10

20

30

40

50

60

70

Incubation Time (Min)

% B

ou

nd

Figure 4.5. Effect of Time of Incubation on In vitro Binding of DLM to Mixture of LDL and

HDL. One µM 14

C-DLM in the mixture of LDL and HDL (50 mg/dL HDL + 100 mg/dL

LDL) was incubated for 15, 30, 60, 120, and 240 min at 37°C in an orbital shaker,

before extracting with isooctane and acetonitrile. Results represent the Mean ± SD (n =

3). There was no statistically significant difference in the % bound DLM compared to 15

min (p>0.05).

Page 164: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

149

2 2.9 4 50.0

2.5

5.0

7.5

10.0

12.5

15.0

17.5

20.0

% HSA

pm

ol/

g b

rain

Figure 4.6. Effect of Binding to Physiological HSA Concentrations on the In vivo Brain

Uptake of DLM. The LCA of adult male SD rats was cannulated and infused (500

µL/min) with 1 µM 14

C-DLM in 2 - 5% HSA solutions for 2 min. Results represent the

Mean ± SD (n = 3 - 4). There was no statistically significant difference in the uptake

among the groups tested (p>0.05).

Page 165: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

150

2.0 2.9 4 50

10

20

30

40

50

60

% HSA

% B

ou

nd

Figure 4.7. In vitro Binding of 1 µM 14

C-DLM to Different Concentrations of HSA (2 –

5%) Following 15 min Incubation at 37°C in an Orbital Shaker. Results represent the

Mean ± SD (n = 3). There was no statistically significant difference in the % bound DLM

among the groups tested (p>0.05).

Page 166: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

151

HBSS HDL LDL LDL + HDL0

10

20

30

40

50p

mo

l/g

bra

in

Figure 4.8. Effect of Binding to Physiological LDL & HDL Concentrations on the In vivo

Brain Uptake of DLM. The LCA of adult male SD rats was cannulated and infused (500

µL/min) with 1 µM 14

C-DLM in 50 mg/dL HDL or 100 mg/dL LDL or their mixture for 2

min. Uptake of 1 µM 14

C-DLM was measured from HBSS buffer alone in the control

group. Results represent the Mean ± SD (n = 3 - 4). There was no statistically significant

difference in the uptake among the groups tested (p>0.05).

Page 167: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

152

HDL LDL LDL+HDL0

10

20

30

40

50

60

70%

Bo

un

d

Figure 4.9. In vitro Binding of 1 µM 14

C-DLM to Physiological Concentrations of LDL

(100 mg/dL), HDL (50 mg/dL) and their Mixture Following 30 min Incubation at 37°C in

an Orbital Shaker. Results represent the Mean ± SD (n = 3). There was no statistically

significant difference in the % bound DLM among the groups tested (p>0.05).

Page 168: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

153

50 100 200 400 0

10

20

30

40

mg/dL

pm

ol/

g b

rain

*

Figure 4.10. Effect of Binding to Lower, Normal and Higher Physiological LDL

Concentrations on the In vivo Brain Uptake of DLM. The LCA of adult male SD rats was

cannulated and infused (500 µL/min) with 1 µM 14

C-DLM in 50, 200 & 400 mg/dL of LDL

solutions respectively for 2 min. Results represent the Mean ± SD (n = 3 - 6). (*)

represents statistically significant differences (p<0.05) compared to 50 mg/dL.

Page 169: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

154

50 100 200 4000

10

20

30

40

50

60

70

80

90

mg/dL

% B

ou

nd

*

Figure 4.11. In vitro Binding of 1 µM 14

C-DLM to Different Concentrations of LDL (50 -

400 mg/dL) Following 30 min Incubation at 37°C in an Orbital Shaker. Results represent

the Mean ± SD (n = 3). (*) represents statistically significant differences (p<0.05) in the

% bound DLM compared to 50 mg/dL.

Page 170: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

155

PND 15 PND 21 Adults0.0

2.5

5.0

7.5

10.0

12.5

15.0

17.5

pm

ol/g

bra

in

Figure 4.12. Effect of Binding to Rat Plasma of Different Age Groups on In vivo Brain

Uptake of DLM. The LCA of adult male SD rats was cannulated and infused (500

µL/min) with 1 µM 14C-DLM in rat plasma of PND 15, PND 21 and PND 90 for 2 min.

Results represent the Mean ± SD (n = 3 - 4). There was no statistically significant

difference in the uptake among the groups tested (p>0.05).

Page 171: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

156

0 - 1 wk 1 wk - 4 wk 4 wk - 1 yr 1 - 3 yrs Adults0

5

10

15

20

25

Age

pm

ol/

g b

rain

Figure 4.13. Effect of Binding to Human Plasma of Different Age Groups on In vivo

Brain Uptake of DLM. The LCA of adult male SD rats was cannulated and infused (500

µL/min) with 1 µM 14C-DLM in human plasma of corresponding age groups for 2 min.

Results represent the Mean ± SD (n = 3 - 5). There was no statistically significant

difference in the uptake among the groups tested (p>0.05).

Page 172: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

157

1.00 10.00 50.000

250

500

750

1000

1250

1500

Adults

PND 21

PND 15

M

pm

ol/

g b

rain

* #

#

#*

*

*

*

Figure 4.14. Effect of Maturity of BBB on In vivo Brain Uptake of DLM. The LCA of PND

15, PND 21 and PND 90 SD rats was cannulated and infused (500 or 250 µL/min) with

1, 10 & 50 µM 14C-DLM in 4% HSA solution for 2 - 4 min. A flow rate of 250 µL/min and

an infusion time of 4 minutes was employed in 15-day and 21-day old rats. Results

represent the Mean ± SD (n = 2 - 8). (*) denotes statistically significant differences

(p<0.05) compared to adults and (#) denotes statistically significant differences (p<0.05)

compared to PND 21.

Page 173: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

158

Conc (µM) PND 15 vs Adults PND 21 vs Adults PND 15 vs PND 21

1 3.7 1.7 2.1

10 2.2 1.5 1.5

50 2.5 1.7 1.5

Table 4.1. Relative Increase in the Brain Uptake of Different Concentrations of DLM in

PND 15 vs PND 21 vs Adults.

Page 174: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

159

0 10 20 30 40 50 600

250

500

750

1000

1250

1500

1750

Adults

PND 21

PND 15

M

pm

ol

/g b

rain

Figure 4.15. Concentration Dependent Uptake of DLM in PND 15, PND 21, and Adult

SD Rats. The LCA of PND 15, PND 21 and PND 90 SD rats was cannulated and

infused (500 or 250 µL/min) with 1, 10 & 50 µM 14C-DLM in 4% HSA solution for 2 - 4

min. A flow rate of 250 µL/min and an infusion time of 4 minutes was employed in PND

15 and PND 21 rats. Results represent the Mean ± SD (n = 2 - 8). Same data as Fig

4.14 but plotted differently.

Page 175: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

160

250 L/min 500 L/min0.0

2.5

5.0

7.5

10.0

12.5

15.0p

mo

l/g

bra

in

Figure 4.16. Effect of Flow Rate on In vivo Brain Uptake of DLM. The LCA of adult male

SD rats was cannulated and infused with 1 µM 14C-DLM in 4% HSA @ 250 µL/min for 4

min. Results represent the Mean ± SD (n = 3). There was no statistically significant

difference in the uptake among the groups tested (p>0.05).

Page 176: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

161

CHAPTER 5

SUMMARY

Despite published data describing the toxicokinetics of pyrethroids in rats, no one

has characterized the systemic distribution of pyrethroids at steady-state. In vivo

tissue:plasma partition coefficients (PCs) reported previously in the literature for

pyrethroids have been based on experimental time-course data from the single dose

studies. The tissue:blood PCs were calculated as the ratio of the tissue AUC to the

blood AUC. These toxicokinetic studies were often conducted at relatively high doses,

which may not be environmentally relevant. Also, the major drawback is that the PC

values reported were not measured at steady-state. To our knowledge, no studies have

addressed whether tissue distribution and PCs of pyrethroids vary significantly during

maturation and development. The series of studies presented here attempted to bridge

this gap in knowledge by characterizing age-dependent differences in the distribution of

pyrethroids in Sprague-Dawley (SD) rats. The overall hypothesis of this work was that

the distribution of pyrethroids in immature rats will be significantly different from that in

adults. To address this hypothesis, we first conducted pilot studies to determine the time

it takes for the plasma levels to reach steady-state with our model system. Steady-state

was attained in adult and in PND 15 and PND 21 rats between 48- and 72-h of constant

infusion using an Alzet pump®. It is generally held that steady-state will be reached

within ≤ 5 half-lives (t½) of a chemical. For DLM, CIS & TRANS, the t½ was reported to

Page 177: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

162

be around 13-h. We first conducted experiments to measure steady-state in vivo

tissue:plasma PCs of DLM, CIS and TRANS in adult male SD rats. For all three

compounds, adipose tissue exhibited much higher PC values than brain, liver, and

skeletal muscle. This is not surprising since fat would be expected to accumulate large

amounts of highly lipophilic pyrethroids (Log P = 4.6 - 6.1). Unexpectedly, brain

exhibited relatively low PCs, as did liver. Skeletal muscle PCs were somewhat higher

compared to brain and liver. At the given dose (Ko = 0.36 mg/h and LD = 150 mg/kg),

we could not detect TRANS in the liver tissue, as the levels were below LOQ. We

postulated that the rapid hydrolysis of TRANS by plasma and liver carboxylesterases

(CEs) in rats were responsible for this observation.

In the second set of studies conducted to assess the influence of maturation on

the systemic distribution of pyrethroids, we determined in vivo tissue:plasma PCs of

DLM, CIS and TRANS in PND 15 and PND 21 pups. For all three pyrethroids, the

tissue:plasma PCs were highest for PND 15 pups as compared to adults, with very few

exceptions. Diminished capacity to metabolize pyrethroids in this age group results in

higher plasma levels, which can saturate relatively low plasma protein levels. Thus, high

circulating levels of pyrethroids combined with lack of adipose tissue results in higher

tissue:plasma PCs in this age group. Greater permeability of the BBB to pyrethroids

results in greater brain:plasma PC in PND 15 pups. The differences in tissue:plasma

PCs become less pronounced between PND 21 and adult rats. De Zwart et al. (2008)

reported that CE activity in both liver and plasma, and CYP 1A1 & 3A2 activity in liver,

reaches adult levels by day 26. CIS tissue:plasma PCs for PND 21 pups were

significantly higher than for adults, probably due to relatively slower metabolism by CYP

Page 178: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

163

450 mediated oxidation resulting in higher plasma levels saturating the protein binding.

Conversely, DLM and TRANS were rapidly metabolized by CE mediated hydrolysis

resulting in relatively lower circulating levels. Sethi et al. (2015) reported that there was

no significant difference in the plasma protein binding of DLM, CIS and TRANS between

PND 21 pups and adult rats. Thus, rapid hydrolysis combined with increased plasma

protein binding in PND 21 pups resulted in tissue:plasma PCs for DLM and TRANS that

were either comparable or lower than in adults. Finally, these data support the

hypothesis that the distribution of pyrethroids in younger rats will be significantly

different from that in adults.

The neurotoxicity of high acute doses of pyrethroids is well characterized.

Accurate measurement of brain uptake is vital to predict target organ dosimetry. To our

knowledge, few studies have been conducted to characterize uptake of pyrethroids into

the brain. Highly lipophilic pyrethroids are expected to diffuse freely across the BBB.

However, previous toxicokinetic studies have reported that peak brain concentrations of

DLM were only about 20% of plasma values. Also, brain:plasma PC value at steady-

state was measured as 0.2 from our PC studies in adult rats. This suggests that

parameters such as plasma protein binding, efflux membrane transporters such as p-gp

may govern the transport of DLM across the BBB. No one has yet illustrated the effect

of these parameters on the brain uptake of DLM. We evaluated this gap of knowledge

by selectively investigating the role of plasma protein binding, membrane transporters

and BBB in limiting the entry of DLM into the brain.

We first confirmed the effect of free fraction on the brain uptake of DLM. We

measured the brain uptake of DLM from 0.01% and 4% HSA using an in situ brain

Page 179: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

164

perfusion technique. We found that the uptake from 0.01% was significantly higher than

that from 4%. Then, we investigated the role of BBB in limiting the entry of DLM into the

brain and obtained exciting results. We disrupted the BBB by prior treatment of mannitol

and then measured the uptake of DLM from HBSS buffer and 4% HSA in control and

mannitol pre-treated animals. Interestingly, even upon the disruption of BBB, the uptake

from 4% HSA did not differ significantly from that of control. In the absence of 4% HSA,

the uptake from HBSS buffer increased significantly in mannitol treated animals. The

BBB thus appears to play some role in opposing the entry of DLM into the brain. In

addition to the BBB, plasma protein binding is also a key player in limiting brain

association of DLM and thus, plays a significant protective role under normal

physiological conditions. We then investigated the role of membrane transporters in

facilitating or inhibiting brain uptake of DLM. We measured the uptake of DLM from

0.01% and 4% HSA in the presence and absence of CSA. Surprisingly, CSA reduced

DLM uptake from 0.01% HSA significantly, while uptake from 4% HSA was not

significantly altered. This finding illustrates that some unidentified influx transporters

contribute to the influx of free DLM across the BBB at high concentrations. These

transporters apparently do not play a role under normal physiological conditions, where

a passive, non-saturable permeation predominates. This data also demonstrates a

significant finding that under physiological albumin concentrations and at low exposure

levels, ontogenic differences in membrane transporters do not affect the brain transport

of DLM. Since CSA is a promiscuous inhibitor of transporters, it was not possible to

identify and characterize a specific transporter family or isoform involved in the

Page 180: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

165

transport. This will require further research. These findings concurred with previous in

vitro data (Appendix Fig. 2B).

Results from PC studies have indicated that the partitioning of pyrethroids into

younger rat’s brain is significantly higher than in adult rats. In general, newborns and

young infants have lower plasma protein binding as compared to adults. Sethi et al.

(2015) reported that the plasma protein binding of pyrethroids in PND 15 pups was

significantly lower than in adult rats. Relatively lower plasma protein binding results in

higher free fractions, which can readily cross the BBB. Also, in immature animals and

neonates, the BBB is not fully functional and is characterized by leaky endothelial

junctions. Therefore, we attempted to assess the influence of age-dependent changes

in plasma protein binding and BBB integrity on the brain uptake of pyrethroids.

We first assessed the potential influence of maturation of plasma protein binding

on the brain entry of DLM, again using the in situ perfusion model. We measured the

uptake of DLM from 2%, 2.9%, 4%, and 5% HSA and found no significant difference

among these groups. This finding was not surprising because there was not a

significant difference in the free fraction of DLM in this range of protein concentrations.

Here, 2.9% and 4% HSA correspond to albumin concentrations in neonates and adult

humans, respectively. These results provided us an insight that age-dependent

differences in plasma protein binding of DLM might not be sufficient to significantly

affect its uptake. We subsequently measured brain uptake of DLM from PND 15, PND

21 and adult rat plasma, and found no significant difference in uptake. We also

measured the uptake of DLM from human pediatric plasma from ages birth – 1 week, 1

– 4 weeks, 4 weeks – 1 year, 1 – 3 years, and adults. As with rat plasma, there was no

Page 181: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

166

significant difference in the uptake of DLM among these groups. The difference in the

free fraction of DLM among the above groups tested was not sufficiently different to

alter brain deposition.

We also measured brain uptake of DLM from physiological concentrations of LDL

(100 mg/dL), HDL (50 mg/dL), and their mixture, in order to characterize their influence

on binding and ensuing brain uptake. Interestingly, the uptake did not vary significantly

among these groups. Not surprisingly, the free fraction of DLM did not differ significantly

either among the groups tested. Under conditions with the same free fraction of DLM,

uptake from physiological concentrations of individual lipoprotein constituents was far

higher than that from 4% HSA. This might be attributed to the presence of lipoprotein

receptors on the surface of brain capillary endothelial cells. It is also possible that DLM

has a higher binding affinity for albumin than lipoprotein constituents due to simple

partitioning into hydrophobic regions of lipoproteins. We also measured the brain uptake

of DLM from different LDL concentrations (50 – 400 mg/dL) to characterize potential

effects on brain uptake in individuals with hypo- and hyper-cholesterolemia. Clearly,

lowest uptake was observed from the highest LDL concentration tested.

The final set of experiments were conducted to study permeability of the

immature BBB to DLM and its role in brain uptake. We measured the uptake of 1, 10

and 50 µM DLM from 4% HSA in PND 15, PND 21 and adult rats using the in situ

perfusion model. The nature of this particular study focused on the relationship between

the BBB penetrability and the brain uptake of DLM, while keeping the free fraction

constant at physiological conditions. At all concentrations tested, the brain uptake of

DLM in PND 15 pups was significantly higher than that of PND 21 and adult rats. Also,

Page 182: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

167

the uptake in PND 21 pups was much higher as compared to adults. Studies have

shown that the rat BBB was not fully developed structurally and was not fully functional

until about the time of weanling. Interendothelial junctions in the immature BBB are

characterized by membrane separations greater than 20 nm and are relatively leaky.

Such junctions in developing rat brain could result in high permeability to DLM. Finally,

these findings support our hypothesis that the brain uptake of pyrethroids in immature

rats will be significantly different from that in adult rats due to differences in plasma

protein binding and BBB permeability. However, we should be very careful while

extrapolating these findings in rats to humans, since the functional maturation of the

BBB as a barrier to substances in the blood appears to occur earlier in humans than

rats.

In conclusion, studies described in this dissertation filled gaps in knowledge that

exist in understanding the differences in the distribution of pyrethroids in immature and

adult rats. For the most part, our results clearly showed that the steady-state

tissue:plasma PCs for pyrethroids were inversely proportional to age. This was

particularly true for PND 15 pups, which exhibited far higher PCs than adults. A few

exceptions were observed in PND 21 pups. The knowledge gained from these studies

can be used for risk assessment of pyrethroids in human sub-populations (adults vs

neonates). Other researchers in the future can refer to these findings to understand

age-dependent differences in the disposition of other pesticides which have similar

physicochemical properties.

In situ brain perfusion studies in this dissertation facilitated reliable measurement

of the uptake of DLM into the brain. The data from these studies demonstrated that the

Page 183: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

168

brain uptake of DLM in the youngest rats is significantly higher than that of mature rats.

However, at low exposure levels, such differences in the uptake were not the result of

ontogenic differences in the expression of membrane transporters or plasma protein

binding. Instead, a more permeable BBB in maturing rats was the reason for

significantly higher brain uptake in this age group. The data obtained from these studies

can be useful in predicting the brain dosimetry of pyrethroids in immature human sub-

populations.

Future studies should be conducted to expound upon the findings presented

here. Since the distribution of pyrethroids is associated with its tissues’ lipid content, it

would be interesting to study the distribution of these highly lipophilic compounds in an

obese rat model. Obesity is often associated with hyperlipidaemia and increased

triglyceride and fat mass. Due to these reasons, distribution of pyrethroids in these

subpopulations may be quite different from that in lean populations. Additionally, based

on our findings related to the contribution of transporters towards the brain uptake of

DLM, a P-gp knockout rat model can be used in the future to definitively demonstrate

that DLM or other pyrethroids are not substrates of this major transporter. The same

technology can be used to identify specific transporter that contribute to the movement

of pyrethroids across the BBB. Studies that characterize the role of lipoprotein receptors

in facilitating the transport of pyrethroids across the BBB can be very informative as

well.

Page 184: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

169

APPENDIX

PLASMA PROTEIN BINDING LIMITS THE BLOOD BRAIN BARRIER

PERMEABILITY OF THE PYRETHROID INSECTICIDE, DELTAMETHRIN4

4Reprinted here with permission from Elsevier. This is an author-produced PDF of an article published in Toxicology Letters following peer review. This manuscript version is made available under the CC-BY-NC-ND 4.0 license: http://creativecommons.org/licenses/by-nc-nd/4.0/ The version of record Manoj A., Anshika S., Jing P., Srinivasa M., Brian S.C., Catherine A.W., James V.B. and Jason Z. (2016) Toxicology Letters. 250-251; 21-28. is available online at: http://www.sciencedirect.com/science/article/pii/S037842741630042X

Page 185: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

170

ABSTRACT

Previous pharmacokinetic studies of deltamethrin (DLM) have revealed that brain levels

of this highly lipophilic pyrethroid insecticide are only 15-20% of plasma levels.

Experiments were performed to assess determinants limiting CNS access including

plasma protein binding and the efflux transporter, P-gp. A human brain microvascular

endothelial cell line, hCMEC/D3, was utilized as a model in vitro system to evaluate

blood-brain barrier (BBB) permeation. Incubation of DLM with a series of human serum

albumin (HSA) concentrations showed that unbound (fu) DLM ranged from 80% with

0.01% HSA to ~20% at the physiologically-relevant 4% HSA. A positive correlation

(R=0.987) was seen between fu and cellular uptake. Concentration-dependent uptake of

DLM in 0.01% HSA was non-linear and was reduced at 4°C and by the P-gp inhibitor

cyclosporine (CSA), indicative of a specific transport process. Cellular accumulation of

[3H]-paclitaxel, a P-glycoprotein (P-gp) substrate, was increased by CSA but not by

DLM, suggesting that DLM is neither a substrate nor an inhibitor of P-gp. The

concentration-dependent uptake of DLM from 4% HSA was linear and not significantly

impacted by temperature or CSA. In situ brain perfusion studies monitoring brain

association of DLM at 0.01% and 4% HSA confirmed the aforementioned in vitro

findings. This study demonstrates that brain uptake of DLM under normal physiological

conditions appears to be a passive, non-saturable process, limited by the high protein

binding of the pyrethroid.

KEYWORDS: Blood-Brain Barrier, Insecticide, Pyrethroid, P-glycoprotein, Transport

Page 186: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

171

1.0 INTRODUCTION

Pyrethroids are the most frequently utilized insecticides in the U.S., since the

phase out of organophosphates (Williams et al., 2008). Pyrethroids are used outdoors in

many agricultural, urban construction and landscaping settings, as well as indoors in

homes and other structures. Certain pyrethroids, such as permethrin, are used to treat

ticks, mites and lice on pets and humans (Frankowski et al., 2010). Thus, it is not

surprising that large segments of the population are exposed to this class of pesticide.

In fact, 3-phenoxybenzoic acid, a metabolite common to many pyrethroids, was

detected in the urine of the majority of over 5,000 persons monitored in the general U.S.

population (Barr et al., 2010). Pyrethroid exposures most frequently result from the

inadvertent ingestion of dusts, hand-to-mouth activity with pets and consumption of very

low levels in foods (Lu et al., 2010; Morgan, 2012).

Although most pyrethroids exert relatively low mammalian toxicity, high doses

can be acutely neurotoxic. Their primary mechanisms of action are interference with the

closure of neuronal voltage-gated calcium and sodium channels (Cao et al., 2011;

Soderlund, 2012). The duration of the closure delay and modification of sodium currents

by deltamethrin (DLM) may last for several seconds, resulting in stimulus-dependent

nerve depolarization and blockage. This results in typical toxic signs of salivation and

hyperexcitability, possibly progressing to choreoathetosis (i.e., CS syndrome).

It would be anticipated that these highly lipophilic compounds should readily

enter and accumulate in the brain. However, DLM peak brain concentrations and areas

under plasma concentration-time curves were found to be only 15-20% of plasma and

blood values (Kim et al., 2008; 2010). This suggests that the CNS deposition of DLM is

Page 187: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

172

governed by other parameters that are not well understood. It has been well established

that binding to albumin and other plasma proteins can significantly reduce the free

fraction (fu) of drugs available for absorption into the CNS. In situ brain extraction of a

series of benzodiazepines in rats, for example, is significantly influenced by their extent

of plasma binding (Jones et al., 1988; Lin and Lin, 1990). Metabolism and systemic

clearance also reduce the amount of free chemical available for CNS uptake. DLM and

other pyrethroids are extensively oxidized by rat and human hepatic microsomal

cytochrome P450s and hydrolyzed by hepatic and plasma carboxylesterases (Anand et

al., 2006; Ross et al., 2006; Scollon et al., 2009). Transport mechanisms are prevalent

within BBB endothelial cells, including a number of Solute Carrier and ATP Binding

Cassette transporters. P-gp, an ATP-dependent efflux transporter, is located on the

apical membrane of endothelial cells of the BBB. Although recent studies from our

laboratory demonstrated that P-gp does not appreciably limit DLM intestinal

permeability using the Caco-2 cell line model (Zastre et al., 2013), it is unknown if P-gp

limits CNS deposition via the BBB. To our knowledge, no one has evaluated potential

determinants of flux for DLM or other pyrethroids across the BBB.

The overall objective of this investigation was to gain an understanding of the

transfer of the pyrethroid, DLM, from the blood into the brain. Emphasis was placed on

clarifying the role of processes generally believed to limit access, in particular, protein

binding and whether it is a substrate for influx or efflux transporters, notably P-gp. A

human brain microvessel endothelial cell line, hCMEC/D3, was selected as a model in

vitro system. These cells display many characteristics of brain endothelium in vivo,

including tight junction formation and expression of transporters (Weksler et al., 2005;

Page 188: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

173

Zastre et al., 2009). This in vitro model was supplemented by in situ brain DLM uptake

experiments in rats.

2.0 MATERIALS AND METHODS

2.1 Materials: Cell culture media and supplements, which include EBM-2 media and

vascular endothelial growth factor, insulin-like growth factor 1, epidermal growth factor,

fibroblast growth factor, hydrocortisone, ascorbate, and gentamycin supplements, were

obtained from Lonza (Allendale, NJ). Rat tail collagen type 1 and trypsin/EDTA were

purchased from BD Biosciences (San Jose, CA) and Mediatech (Manassas, VA),

respectively. Fetal bovine serum (FBS), mannitol, and human serum albumin (HSA)

were purchased from Sigma-Aldrich (St. Louis, MO). Cell culture flasks, plates, and

dishes were obtained from Greiner Bio-one (Monroe, NC). Cyclosporine A (CSA) was

purchased from Amresco (Salon, OH). Radiolabeled [14C]-DLM (54.1 mCi/mmol) was

kindly donated by Bayer CropScience (Research Triangle Park, NC). [3H]-Paclitaxel

(PTX) (23 Ci/mmol) was obtained from Moravek Biochemicals (Brea, CA), while iso-

octane and acetonitrile were purchased from EMD Chemicals (Billerica, MA).

2.2 Cell Culture: hCMEC/D3 cells were kindly donated by Dr. Babette Weksler, Weill

Cornell Medical College. Cells were maintained at 37ºC, 5% CO2, and 95% humidified

air in EBM-2 media supplemented with vascular endothelial growth factor, insulin-like

growth factor-1, epidermal growth factor, fibroblast growth factor, hydrocortisone,

ascorbate, gentamycin, and 2.5% FBS as previously described (Weksler et al., 2005).

Cells were seeded onto rat-tail collagen type-1 coated flasks and 24-well plates.

Page 189: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

174

2.3 Protein Binding of DLM: Protein binding of DLM with HSA was measured using a

modification of a procedure previously reported (Sethi et al., 2014). Briefly, 1.0 μM [14C]-

DLM was incubated with increasing concentrations of HSA (0.01, 0.1, 1.0, and 4.0%

w/v) dissolved in PBS at 370C for 15 min. Two volumes of iso-octane were added and

vortexed for 1 min, followed by centrifugation at 14,000xg for 5 min to separate aqueous

and organic layers. The organic layer was removed, and the free (unbound) fraction it

contained was quantified using a liquid scintillation counter (LS 6500, Beckman Coulter,

Brea, CA). The protein-bound fraction was quantified by subjecting the aqueous layer to

protein precipitation using 3 volumes of acetonitrile. The sample was vortexed for 1 min

and centrifuged at 14,000xg for 5 min. The supernatant was removed and quantified by

liquid scintillation counting. To establish the effect of DLM concentration on protein

binding, increasing concentrations of [14C]-DLM ranging from 0.1 – 10 μM with either

0.01% or 4.0% HSA was assessed as described above.

2.4 Cellular Uptake Experiments: The uptake of 1.0 M [14C]-DLM by hCMEC/D3 cells

from a series of HSA concentrations (0.01, 0.1, 1.0, and 4.0% w/v) was assessed at

37ºC for 3 min (Linear range of uptake – Suppl. Fig. 1). Prior to uptake, the cells were

washed twice with pre-warmed HBSS (Mediatech) containing 10 mM HEPES pH = 7.2

(hereafter, referred to as transport buffer), and pre-incubated for 15 min with transport

buffer. Subsequently, 1.0 μM [14C]-DLM in HSA containing transport buffer was added

to the wells. After the uptake time, the cells were washed 3 times with ice-cold PBS and

lysed (1% Triton X-100, 50 mM Tris, 250 mM NaCl pH=8.0). The amount of DLM was

quantified using liquid scintillation counting, and the results were normalized to total

protein using a BCA protein assay kit (Thermo Scientific, Rockford, IL).

Page 190: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

175

A similar protocol was used to establish whether DLM is a substrate or inhibitor

for P-gp-mediated efflux. Inhibitor properties were assessed by determining the uptake

of the P-gp substrate [3H]-PTX (4 nM) with or without the P-gp inhibitor, CSA (25 μM),

and DLM (10 μM) at 370C for 30 min in transport buffer. For substrate properties of DLM

for P-gp, the uptake of 1.0 M [14C]-DLM was monitored in the presence of the P-gp

inhibitor CSA (25 μM) from 0.01% and 4% HSA (low and high protein binding extremes,

respectively). The amount of PTX or DLM was quantified using liquid scintillation

counting and normalized to total protein.

2.5 Kinetic Analysis of DLM Transport: The concentration-dependent uptake of DLM

was determined to evaluate its transport kinetics for estimation of the Michaelis–Menton

constant (Km) and maximal transport velocity (Vmax) in hCMEC/D3 cells. The uptake (3

min) of increasing concentrations of [14C]-DLM (0.1-10 μM) in transport buffer containing

0.01% or 4% HSA was performed as described above. The total uptake rate into

hCMEC/D3 cells is the sum of all saturable (specific; nonlinear) and non-saturable

(nonspecific; linear) components. Two approaches were utilized to estimate the non-

saturable contribution including measuring the concentration-dependent uptake of DLM

at 4ºC or in the presence of 25 μM CSA (37ºC uptake). Both were included since over

estimation may be associated with reduced temperatures decreasing passive diffusion

and membrane fluidity (Poirier et al., 2008). The concentration of DLM for the analysis

was adjusted based on the free fraction available for uptake that was determined as

described above. Kinetic analysis was performed by applying a one-site Michaelis–

Menten equation with a nonsaturable component using Graphpad Prism 6 software.

Page 191: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

176

Equation 1: 𝑉 =𝑉max[𝑆]

𝐾m+[𝑆]+ 𝑘ns[𝑆]

Where V is the total rate of uptake, Vmax is the maximum uptake rate, Km is the

Michaelis-Menton constant, [S] is the substrate concentration, and kns is the coefficient

for nonspecific uptake by diffusion.

2.6 In Situ Brain Perfusion:

2.6.1 Animal surgical preparation: Brain-associated DLM levels were

measured using a modified in situ rat brain perfusion technique, originally developed by

Takasato et al (Boje, 2001; Takasato et al., 1984). The University of Georgia Animal

Care and Use Committee approved the protocol for this study. Male Sprague-Dawley

(SD) rats (300-350g) were purchased from Charles River Laboratories (Raleigh, NC).

Each rat was housed in a cage with a 12-h light/dark cycle at ambient temperature

(22ºC) and relative humidity (55 ± 5%). Food (5001 Rodent Diet, PMI Nutrition LLC,

Brentwood, MO) and water were provided ad libitum. Rats were anesthetized with KAX

(Ketamine, Acepromazine, and Xylazine) cocktail. The left common carotid artery was

exposed and cannulated with a 25G needle affixed to PE-50 tubing for perfusion fluid

administration. Prior to the start of the infusion, the heart was stopped by rapidly

severing the cardiac ventricles to eliminate contributions of vascular perfusion from the

systemic circulation. The left common carotid artery was then perfused with 2 mL of

sterile saline to flush blood from the brain. This was followed by the infusion of

corresponding perfusion fluids.

Since the perfusion fluid was infused into the left common carotid artery, only the

left cerebral hemisphere was processed for DLM quantitation. The isolated brain tissue

Page 192: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

177

was homogenized with a Tekmar Tissumizer IKA Ultra-Turrax homogenizer (Janke and

Kunkel Laboratories, Cridersville, OH), by adding two volumes of ice-cold distilled water

by weight. An aliquot (100 µL) of the brain homogenate was added to 4 mL of EcoLite

scintillation cocktail (MP Biomedicals, Solon, OH). Uptake of DLM was quantified by

measuring the radioactivity in each sample by liquid scintillation counting using a

Beckman Coulter LS 6500 (Brea, CA) and normalizing to tissue weight.

2.6.2 Effect of HSA and CSA on DLM associated brain levels: The perfusion

fluid consisted of 10 µM [14C]-DLM and 0.01 or 4% HSA with or without the P-gp

inhibitor CSA (25 µM) in HBSS buffer. All solutions were incubated for 15 min at 37°C

prior to the infusion. In the CSA group, animals were pre-treated with CSA prior to the

start of DLM infusion by perfusing 2 mL of sterile saline containing 25 µM CSA. The

DLM containing perfusion fluid (including 25 µM CSA) was then infused at a constant

rate of 500 µL/min for 2 min using a Harvard 22 syringe infusion pump (Harvard

Apparatus, Holliston, MA).

2.6.3 Role of the BBB in limiting DLM associated brain levels: Mannitol (1.6

M) was infused at 0.25 ml/kg/sec for 30 seconds prior to the perfusion of DLM to disrupt

the BBB (Cosolo et al., 1989). The perfusion fluid consisted of 1 µM [14C]-DLM with or

without 4% HSA in HBSS buffer was then infused at a constant rate of 500 µL/min for 2

min using a Harvard 22 syringe infusion pump.

2.7 Statistical Analysis. All experiments were performed with a minimum of three

independent experiments unless otherwise stated. Statistical significance was evaluated

between groups using either Student’s t-test or one way ANOVA, followed by Tukey’s

Page 193: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

178

multiple comparison test, as indicated, with a significance level of p<0.05, using

Graphpad Prism 6 software® (San Diego, CA).

3.0 RESULTS

3.1 Impact of Protein Binding on DLM Cellular Uptake

Protein binding experiments were performed with increasing concentrations of HSA to

establish the free fraction of DLM. The percent DLM unbound decreased with increasing

concentration of HSA ranging from ~80% with 0.01% HSA to ~20% at the

physiologically-relevant concentration of 4% HSA (Fig. 1A). The free fraction of DLM

(~80%) with 0.01% HSA was unchanged over a range of DLM concentrations (Fig. 1B).

At 4% HSA, an increase in the free fraction from ~20% to ~40% was observed only at

high concentrations of DLM ranging from 1 to 10 µM (Fig. 1C). To establish whether a

relationship exists between the extent of DLM uptake by hCMEC/D3 cells and the free

fraction, cellular uptake of DLM from increasing concentrations of HSA was determined.

Figure 1D demonstrates a progressive decrease in DLM uptake by hCMEC/D3 cells

with increasing concentration of HSA. A strong correlation (R=0.987) was found

between the free fractions of DLM and the extent of DLM uptake by hCMEC/D3 cells

(Fig. 1E).

3.2 Role of P-glycoprotein

The functional activity of P-gp in our hCMEC/D3 model system was demonstrated by an

increased cellular accumulation of the P-gp substrate PTX in the presence of the P-gp

inhibitor CSA (Fig. 2A). In contrast, DLM had no effect on PTX accumulation. To

elucidate the potential role of P-gp in limiting DLM accumulation in the brain, DLM

Page 194: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

179

uptake was assessed in the presence of 25 µMCSA. The uptake of DLM was

significantly reduced by CSA in the presence of 0.01% HSA, corresponding to a high

free fraction of DLM (Fig. 2B). However, CSA had no effect on DLM uptake in the

presence of 4% HSA (low DLM free fraction) (Fig. 2B). The presence of CSA had no

effect on the extent of DLM protein binding with either 0.01% or 4% HSA (Suppl. Fig.

2).

3.3 Concentration-Dependent Uptake of DLM

To establish the transport kinetics, the concentration-dependent uptake of DLM by

hCMEC/D3 cells was assessed at the two extremes of protein binding/free fraction (i.e.

0.01% and 4% HSA). Although the concentrations evaluated ranged from 0.1 to 10 M,

Fig. 3 represents only the available free fraction of DLM at each concentration that was

calculated based on Fig. 1B and C.

The cellular uptake of DLM from 4% HSA (low free fraction) demonstrated linear

kinetics (Fig. 3A). In contrast, DLM exhibited non-linear kinetics with 0.01% HSA (high

free fraction) (Fig. 3B). Furthermore, the extent of DLM uptake was greater from 0.01%

HSA than 4% HSA. The concentration-dependent uptake of DLM was evaluated at 4ºC

to reduce energy-dependent processes in the cell. Uptake of DLM at 4ºC was

substantially lower than at 37ºC and linear over the DLM concentration range with

0.01% HSA (high free fraction) (Fig. 3B). No difference was observed for DLM uptake

at 4ºC and 37ºC with 4% HSA (low free fraction) (Fig. 3A). Similarly, the inclusion of

CSA had no effect on DLM uptake from 4% HSA, but reduced uptake over the

concentration range in the presence of 0.01% HSA.

Page 195: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

180

The linearity of DLM transport with 0.01% HSA at both 40C and with the inclusion

of CSA allows for an estimation of the non-saturable (passive) uptake component. To

estimate the saturable transport kinetics of DLM, equation 1 was fitted to the total and

non-saturable uptake components. A saturable curve (dotted line) was obtained for

DLM with Vmax of 327 +/- 12.2 pmol/mg protein/min and Km of 2.3 +/- 0.3 M using 4°C

for the non-saturable component (Fig. 3C). Similar values were obtained using CSA

(solid line) as the non-saturable component with a Vmax of 110 +/- 13.2 pmol/mg

protein/min and a Km of 1.4 +/- 0.3 M.

3.4 In Situ brain perfusion of DLM

The extent of brain associated DLM from a perfusate containing 0.01% HSA (high free

fraction) was significantly greater than from 4% HSA (low free fraction) (Fig. 4A).

Similarly to the in vitro studies, the presence of CSA significantly reduced brain

association of DLM from 0.01% HSA but not from 4% HSA. Fig. 4B illustrates the extent

of brain association of DLM with and without 4% HSA after BBB disruption using

mannitol. In the absence of protein (HBSS group), pre-treatment with mannitol

significantly increased the brain association of DLM compared to control. In the

presence of 4% HSA, the pre-treatment with mannitol did not have a significant impact

on brain association of DLM.

4.0 DISCUSSION

The BBB is an effective regulator of the entry of endo- and xenobiotics into the

brain. There is generally a correlation between the rate at which a compound enters the

brain and its lipophilicity (Clark, 2003; Levin, 1980). Even though DLM is a highly

Page 196: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

181

lipophilic compound (LogP = 6.1), brain levels are relatively low compared to the plasma

compartment (Kim et al., 2008; 2010). Several research groups have described

parabolic relationships between the lipophilicity of drugs and their brain

uptake/pharmacological activity (Dishino et al., 1983; Rowley et al., 1997). Moderately

lipophilic compounds exhibit the highest uptake, but a point of diminishing return is

reached, beyond which higher LogP values result in decreasing BBB penetration. DLM

and other pyrethroids satisfy two criteria for poor membrane permeability, namely

molecular masses >500 and logP > 5 (Lipinski et al., 2001). Non-specific membrane

protein and lipoprotein binding, coupled with lipid partitioning also serve to reduce the

flux of highly-lipid soluble compounds through the BBB, by virtually trapping them there

(Liu et al., 2011; Nau et al., 2010; Waterhouse, 2003).

Another important factor in the extent to which xenobiotics cross the BBB is the

unbound fraction in plasma (Kalvass and Maurer, 2002; Kalvass et al., 2007; Tanaka

and Mizojiri, 1999). Binding to albumin reduces the amount of compound free to

permeate the BBB (Tanaka and Mizojiri, 1999). Since DLM has a high degree (90%) of

protein and lipoprotein binding in plasma (Sethi et al., 2014), only a small fraction

(unbound) of an applied dose would be considered available for transport across

endothelial cells into the brain. Results of the current investigation demonstrate that the

extent of binding of DLM to HSA is an important determinant of disposition by brain

endothelial cells in vitro. It is noteworthy that DLM uptake by hCMEC/D3 cells was quite

low in the presence of 4% HSA, a concentration frequently present in vivo.

In situ brain perfusion is one of the most sensitive and physiologically-relevant

techniques for estimation of CNS uptake of drugs and other chemicals (Bickel, 2005).

Page 197: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

182

The rat brain perfusion model has been successfully used to accurately measure uptake

of a variety of neurotoxic chemicals and neuroactive drugs (Kumar et al., 2007; Rabin et

al., 1993). Rowley et al. (1997) used the model to assess BBB penetration of a series of

anticonvulsants, demonstrating that albumin binding reduced brain uptake, as did

excessive lipid solubility. Similarly, brain extraction increased as the unbound fraction

and lipophilicity increased for a series of benzodiazepines (Jones et al., 1988; Lin and

Lin, 1990). In the present study, uptake of DLM into the brain of rats was significantly

lower from perfusate with a physiological HSA concentration than from perfusate with a

low HSA concentration. These findings are consistent with our in vitro results that

demonstrated greater hCMEC/D3 accumulation with increasing DLM unbound fraction.

Hypertonic mannitol was infused in order to assess the influence of the BBB on

limiting brain uptake of DLM. Electron microscopy has demonstrated that mannitol

disrupts the BBB by reducing endothelial cell size and opening endothelial tight

junctions (Brown et al., 2004; Cosolo et al., 1989). Mannitol infusion produced almost a

3-fold increase in the brain association of DLM in the absence of HSA demonstrating

that the BBB is an effective barrier limiting brain penetration of DLM. In the presence of

4% HSA, brain association of DLM was not significantly altered by mannitol pre-

treatment. Albumin, and correspondingly DLM bound to albumin, is too large a molecule

(Molecular Weight ∼66,000) to cross the BBB, even when the tight junctions between

the endothelial cells are disrupted by hypertonic mannitol (Brown et al., 2004).

Therefore, our findings at 4% HSA illustrate that plasma protein binding limits brain

association of this neurotoxic insecticide and plays a significant protective role under

normal physiological conditions.

Page 198: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

183

Our observations of DLM transport into hCMEC/D3 cells are similar in several

respects to those with the in vitro intestinal transport cell line model, Caco-2 (Zastre et

al., 2013). There was little evidence of P-gp-mediated efflux of DLM and no effect on

accumulation of P-gp substrates in either cell type, indicating that this pyrethroid is not a

P-gp inhibitor or substrate. Kinetic analysis of DLM uptake revealed a nonlinear curve at

a high DLM free fraction in hCMEC/D3 cells that was analogous to the non-linearity

observed in Caco-2 cells in the absence of HSA. CSA was capable of reducing the

uptake of DLM by hCMEC/D3 cells as well as brain uptake using the in situ brain

perfusion model. CSA significantly reduced the brain uptake of DLM from 0.01% HSA,

where higher free fractions are available to interact with the transporters. With 4% HSA,

the free fractions are not high enough for CSA to significantly impact uptake. Although

CSA is a well-established P-gp inhibitor, it is also known to be a promiscuous inhibitor of

both influx and efflux transporters (Letschert et al., 2006; Tang et al., 2002). Inhibition of

DLM uptake by CSA was also found in an intestinal cell line, Caco-2 (Zastre et al.,

2013). The reduction of DLM uptake at high unbound fractions, at 4°C, and in the

presence of CSA are all indicative of a specific influx transport process. Kinetic analysis

of the DLM uptake by hCMEC/D3 cells from 0.01% HSA resulted in a Km of 1.3 M

using 4°C as the non-saturable component, which is similar to the Km in Caco-2 cells of

1.5 M. Both the BBB and the intestinal tract express many of the same transporters,

and the similarity in the Km values suggests a common transporter maybe involved (Sai

and Tsuji, 2004). However, our findings do not identify which transporter family or

isoform may be involved in DLM influx transport. Conversely, DLM uptake from 4%

albumin exhibited linearity, and the uptake at 4°C or with CSA was not inhibited. The

Page 199: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

184

brain uptake of DLM from 4% albumin also exhibited linearity in the in situ brain

perfusion experiment. These in vitro findings corroborated by in situ results reflect a

passive, non-saturable BBB transport process predominating under normal

physiological conditions.

Pronounced variations in albumin concentrations may result in changes in the

fraction of DLM unbound and ultimately in the extent of BBB penetration. Our results

demonstrate that substantial DLM uptake into hCMEC/D3 cells progressively increases

when albumin concentrations are less than 1%. Plasma albumin levels in humans vary

with age, ranging from 3.3% in neonates to 4.5% in adults (Kanakoudi et al., 1995;

Lerman et al., 1984). Although ontogenic differences in the expression of transporters

have been characterized (Daood et al., 2008; de Zwart et al., 2008), marked

displacement of DLM from albumin binding sites and/or disease conditions producing

hypoalbuminemia would likely need to be present for age-dependent differences in

carrier-mediated processes to influence BBB permeability to DLM. Age-dependent

differences in BBB penetration, or DLM uptake by the brain may not be prominent when

there are normal physiological levels of albumin.

An additional variable in DLM sequestration in vivo may be lipoproteins. Sethi et

al. (2014) reported that DLM and permethrin, another highly lipophilic pyrethroid, were

bound by both albumin and lipoproteins. Thus, lipoprotein binding may also be a

significant determinant of the plasma free fraction available for brain uptake. It therefore

appears worthwhile to establish how the binding of DLM to both albumin and

lipoproteins in human plasma may impact brain uptake of DLM, and whether age-

related changes in their plasma concentrations could be of importance. Future studies

Page 200: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

185

will be conducted to measure brain uptake of DLM from human plasma of neonates and

adults, using the in situ rat brain perfusion technique employed in the current

investigation.

In conclusion, our results indicate that the efflux transporter P-gp does not

appreciably contribute to the lower than anticipated accumulation of DLM in the brain.

Although a low affinity transporter is suggested to contribute to DLM uptake by the

brain, this is only significant at high free fractions of DLM not observed under normal

physiological conditions or in younger age groups. The extensive binding and low

penetration of DLM with physiological concentrations of albumin suggest that the

modest brain levels of DLM observed in vivo are due to poor BBB permeation by the

highly lipophilic compound and its limited free fraction available for diffusion.

Acknowledgement: The authors would like to thank the Council for the Advancement

of Pyrethroid Human Risk Assessment (CAPHRA) for funding this work. MA was

partially supported by a University of Georgia Interdisciplinary Toxicology Program

graduate stipend.

Page 201: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

186

REFERENCES

Anand, S.S., Bruckner, J.V., Haines, W.T., Muralidhara, S., Fisher, J.W., Padilla, S.,

2006. Characterization of deltamethrin metabolism by rat plasma and liver microsomes.

Toxicol Appl Pharmacol 212, 156-166.

Barr, D.B., Olsson, A.O., Wong, L.Y., Udunka, S., Baker, S.E., Whitehead, R.D.,

Magsumbol, M.S., Williams, B.L., Needham, L.L., 2010. Urinary Concentrations of

Metabolites of Pyrethroid Insecticides in the General US Population: National Health

and Nutrition Examination Survey 1999-2002. Environ Health Perspect 118, 742-748.

Bickel, U., 2005. How to measure drug transport across the blood-brain barrier.

NeuroRx 2, 15-26.

Boje, K.M., 2001. In vivo measurement of blood-brain barrier permeability. Curr Protoc

Neurosci Chapter 7, Unit 7 19.

Brown, R.C., Egleton, R.D., Davis, T.P., 2004. Mannitol opening of the blood-brain

barrier: regional variation in the permeability of sucrose, but not 86Rb+ or albumin.

Brain Res 1014, 221-227.

Cao, Z., Shafer, T.J., Murray, T.F., 2011. Mechanisms of pyrethroid insecticide-induced

stimulation of calcium influx in neocortical neurons. J Pharmacol Exp Ther 336, 197-

205.

Clark, D.E., 2003. In silico prediction of blood-brain barrier permeation. Drug Discov

Today 8, 927-933.

Page 202: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

187

Daood, M., Tsai, C., Ahdab-Barmada, M., Watchko, J.F., 2008. ABC transporter (P-

gp/ABCB1, MRP1/ABCC1, BCRP/ABCG2) expression in the developing human CNS.

Neuropediatrics 39, 211-218.

de Zwart, L., Scholten, M., Monbaliu, J.G., Annaert, P.P., Van Houdt, J.M., Van den

Wyngaert, I., De Schaepdrijver, L.M., Bailey, G.P., Coogan, T.P., Coussement, W.C.,

Mannens, G.S., 2008. The ontogeny of drug metabolizing enzymes and transporters in

the rat. Reprod Toxicol 26, 220-230.

Dishino, D.D., Welch, M.J., Kilbourn, M.R., Raichle, M.E., 1983. Relationship between

lipophilicity and brain extraction of C-11-labeled radiopharmaceuticals. J Nucl Med 24,

1030-1038.

Frankowski, B.L., Bocchini, J.A., Jr., Council on School, H., Committee on Infectious,

D., 2010. Head lice. Pediatrics 126, 392-403.

Jones, D.R., Hall, S.D., Jackson, E.K., Branch, R.A., Wilkinson, G.R., 1988. Brain

uptake of benzodiazepines: effects of lipophilicity and plasma protein binding. J

Pharmacol Exp Ther 245, 816-822.

Kalvass, J.C., Maurer, T.S., 2002. Influence of nonspecific brain and plasma binding on

CNS exposure: implications for rational drug discovery. Biopharm Drug Dispos 23, 327-

338.

Kalvass, J.C., Maurer, T.S., Pollack, G.M., 2007. Use of plasma and brain unbound

fractions to assess the extent of brain distribution of 34 drugs: comparison of unbound

Page 203: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

188

concentration ratios to in vivo p-glycoprotein efflux ratios. Drug Metab Dispos 35, 660-

666.

Kanakoudi, F., Drossou, V., Tzimouli, V., Diamanti, E., Konstantinidis, T., Germenis, A.,

Kremenopoulos, G., 1995. Serum concentrations of 10 acute-phase proteins in healthy

term and preterm infants from birth to age 6 months. Clin Chem 41, 605-608.

Kim, K.B., Anand, S.S., Kim, H.J., White, C.A., Bruckner, J.V., 2008. Toxicokinetics and

tissue distribution of deltamethrin in adult Sprague-Dawley rats. Toxicol Sci 101, 197-

205.

Kim, K.B., Anand, S.S., Kim, H.J., White, C.A., Fisher, J.W., Tornero-Velez, R.,

Bruckner, J.V., 2010. Age, dose, and time-dependency of plasma and tissue distribution

of deltamethrin in immature rats. Toxicol Sci 115, 354-368.

Kumar, G., Smith, Q.R., Hokari, M., Parepally, J., Duncan, M.W., 2007. Brain uptake,

pharmacokinetics, and tissue distribution in the rat of neurotoxic N-

butylbenzenesulfonamide. Toxicol Sci 97, 253-264.

Lerman, J., Gregory, G.A., Willis, M.M., Eger, E.I., 2nd, 1984. Age and solubility of

volatile anesthetics in blood. Anesthesiology 61, 139-143.

Letschert, K., Faulstich, H., Keller, D., Keppler, D., 2006. Molecular characterization and

inhibition of amanitin uptake into human hepatocytes. Toxicol Sci 91, 140-149.

Levin, V.A., 1980. Relationship of octanol/water partition coefficient and molecular

weight to rat brain capillary permeability. J Med Chem 23, 682-684.

Page 204: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

189

Lin, T.H., Lin, J.H., 1990. Effects of protein binding and experimental disease states on

brain uptake of benzodiazepines in rats. J Pharmacol Exp Ther 253, 45-50.

Lipinski, C.A., Lombardo, F., Dominy, B.W., Feeney, P.J., 2001. Experimental and

computational approaches to estimate solubility and permeability in drug discovery and

development settings. Adv Drug Del Rev 46, 3-26.

Liu, X., Testa, B., Fahr, A., 2011. Lipophilicity and its relationship with passive drug

permeation. Pharm Res 28, 962-977.

Lu, C.S., Schenck, F.J., Pearson, M.A., Wong, J.W., 2010. Assessing Children's Dietary

Pesticide Exposure: Direct Measurement of Pesticide Residues in 24-Hr Duplicate Food

Samples. Environ Health Perspect 118, 1625-1630.

Morgan, M.K., 2012. Children's exposures to pyrethroid insecticides at home: a review

of data collected in published exposure measurement studies conducted in the United

States. Int J Environ Res Public Health 9, 2964-2985.

Nau, R., Sorgel, F., Eiffert, H., 2010. Penetration of drugs through the blood-

cerebrospinal fluid/blood-brain barrier for treatment of central nervous system infections.

Clin Microbiol Rev 23, 858-883.

Poirier, A., Lave, T., Portmann, R., Brun, M.E., Senner, F., Kansy, M., Grimm, H.P.,

Funk, C., 2008. Design, data analysis, and simulation of in vitro drug transport kinetic

experiments using a mechanistic in vitro model. Drug Metab Dispos 36, 2434-2444.

Page 205: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

190

Rabin, O., Hegedus, L., Bourre, J.M., Smith, Q.R., 1993. Rapid brain uptake of

manganese(II) across the blood-brain barrier. J Neurochem 61, 509-517.

Ross, M.K., Borazjani, A., Edwards, C.C., Potter, P.M., 2006. Hydrolytic metabolism of

pyrethroids by human and other mammalian carboxylesterases. Biochem Pharmacol

71, 657-669.

Rowley, M., Kulagowski, J.J., Watt, A.P., Rathbone, D., Stevenson, G.I., Carling, R.W.,

Baker, R., Marshall, G.R., Kemp, J.A., Foster, A.C., Grimwood, S., Hargreaves, R.,

Hurley, C., Saywell, K.L., Tricklebank, M.D., Leeson, P.D., 1997. Effect of plasma

protein binding on in vivo activity and brain penetration of glycine/NMDA receptor

antagonists. J Med Chem 40, 4053-4068.

Sai, Y., Tsuji, A., 2004. Transporter-mediated drug delivery: recent progress and

experimental approaches. Drug Discov Today 9, 712-720.

Scollon, E.J., Starr, J.M., Godin, S.J., DeVito, M.J., Hughes, M.F., 2009. In vitro

metabolism of pyrethroid pesticides by rat and human hepatic microsomes and

cytochrome p450 isoforms. Drug Metab Dispos 37, 221-228.

Sethi, P.K., Muralidhara, S., Bruckner, J.V., White, C.A., 2014. Measurement of plasma

protein and lipoprotein binding of pyrethroids. J Pharmacol Toxicol Methods 70, 106-

111.

Soderlund, D.M., 2012. Molecular mechanisms of pyrethroid insecticide neurotoxicity:

recent advances. Arch Toxicol 86, 165-181.

Page 206: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

191

Takasato, Y., Rapoport, S.I., Smith, Q.R., 1984. An in situ brain perfusion technique to

study cerebrovascular transport in the rat. Am J Physiol 247, H484-H493.

Tanaka, H., Mizojiri, K., 1999. Drug-protein binding and blood-brain barrier permeability.

J Pharmacol Exp Ther 288, 912-918.

Tang, F., Horie, K., Borchardt, R.T., 2002. Are MDCK cells transfected with the human

MDR1 gene a good model of the human intestinal mucosa? Pharm Res 19, 765-772.

Waterhouse, R.N., 2003. Determination of lipophilicity and its use as a predictor of

blood-brain barrier penetration of molecular imaging agents. Mol Imaging Biol 5, 376-

389.

Weksler, B.B., Subileau, E.A., Perriere, N., Charneau, P., Holloway, K., Leveque, M.,

Tricoire-Leignel, H., Nicotra, A., Bourdoulous, S., Turowski, P., Male, D.K., Roux, F.,

Greenwood, J., Romero, I.A., Couraud, P.O., 2005. Blood-brain barrier-specific

properties of a human adult brain endothelial cell line. FASEB J 19, 1872-1874.

Williams, M.K., Rundle, A., Holmes, D., Reyes, M., Hoepner, L.A., Barr, D.B., Camann,

D.E., Perera, F.P., Whyatt, R.M., 2008. Changes in pest infestation levels, self-reported

pesticide use, and permethrin exposure during pregnancy after the 2000-2001 U.S.

Environmental Protection Agency restriction of organophosphates. Environ Health

Perspect 116, 1681-1688.

Zastre, J., Dowd, C., Bruckner, J., Popovici, A., 2013. Lack of P-glycoprotein-mediated

efflux and the potential involvement of an influx transport process contributing to the

Page 207: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

192

intestinal uptake of deltamethrin, cis-permethrin, and trans-permethrin. Toxicol Sci 136,

284-293.

Zastre, J.A., Chan, G.N.Y., Ronaldson, P.T., Ramaswamy, M., Couraud, P.O., Romero,

I.A., Weksler, B., Bendayan, M., Bendayan, R., 2009. Up-regulation of P-glycoprotein by

HIV protease inhibitors in a human brain microvessel endothelial cell line. J Neurosci

Res 87, 1023-1036.

Page 208: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

193

Fig. 1. Relationship between HSA concentration, DLM binding, and uptake of DLM by

hCMEC/D3 cells. (A) Extent of protein binding of 1.0 μM [14C]-DLM with increasing

concentrations of HSA for 15 min at 37ºC. (B) Protein binding with increasing

concentrations of [14C]-DLM at 0.01% HSA for 15 min at 37ºC. (C) Protein binding with

increasing concentrations of [14C]-DLM at 4% HSA for 15 min at 37ºC. (D) Accumulation

of 1.0 M [14C]-DLM by hCMEC/D3 cells with increasing concentrations of HSA at 37ºC

for 3 min. (E) Correlation between the extent of DLM uptake by hCMEC/D3 cells at

varying HSA concentrations with the unbound fraction of DLM. All results are expressed

as the mean +/- SD (n = 3).

Page 209: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

194

Fig. 2. Assessment of the P-gp substrate and inhibitor properties of DLM in hCMEC/D3

cells. (A) Uptake by hCMEC/D3 cells of [3H]-paclitaxel (4 nM) with or without the P-gp

inhibitor, CSA (25 μM), and DLM (10 μM) at 37ºC for 30 min. Results are expressed as

the mean +/- SD (n = 3). () denotes statistically significant differences (p<0.05). (B)

Uptake of 1.0 M [14C]-DLM by hCMEC/D3 cells with 0.01 and 4% HSA at 37ºC for 3

min with and without CSA (25 μM). Results are expressed as the mean +/- SD (n = 3).

() denotes statistically significant differences (p<0.05).

Page 210: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

195

Fig. 3. Concentration-dependent uptake of DLM by hCMEC/D3 cells. (A) Uptake (3 min)

of increasing concentrations of [14C]-DLM in transport buffer containing 4% HSA at ()

37ºC, () 4ºC, and (☐) with 25 µM CSA at 37ºC. Results are expressed as the mean +/-

SD (n=6). (B) Uptake (3 min) of increasing concentrations of [14C]-DLM in transport

buffer containing 0.01% HSA at () 37ºC, () 4ºC, and (☐) with 25 µM CSA at 37ºC.

Results are expressed as the mean +/- SD (n=6). (C) The saturable DLM uptake was

estimated by curve fitting the total (37ºC) and non-saturable uptake components from

4ºC (Dotted line) or from CSA (Solid line).

Page 211: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

196

Figure 4: (A) Effect of HSA and CSA on the in vivo brain association of DLM. The Left

common carotid artery of adult male Sprague-Dawley rats was cannulated and infused

(500 µL/min) with 10 µM [14C]-DLM in 0.01% and 4% HSA solutions either alone or in

combination with 25 µM CSA for 2 min. Results represent the mean ± SD (n = 4 - 7).

() denotes statistically significant differences (p<0.05). (B) Effect of mannitol on the in

vivo brain association of DLM with and without 4% HSA. The Left common carotid

artery of adult male Sprague-Dawley rats was cannulated and infused (500 µL/min) with

1 µM [14C]-DLM with and without 4% HSA. In the mannitol group, animals were pre-

treated with 1.6 M Mannitol prior to infusion of DLM. Results represent the mean ± SD

(n = 4 - 5). () denotes statistically significant differences (p<0.05).

Page 212: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

197

Supplementary Fig. 1. Time dependent accumulation of 1.0 µM [14C]-DLM by

hCMEC/D3 cells with 0.01% HSA at 37ºC. Results are expressed as the Mean ± SD (n

= 3).

Page 213: CHARACTERIZATION OF DISPOSITION OF DELTAMETHRIN, CIS

198

Supplementary Fig. 2. (A) Effect of CSA (25 µM) on the protein binding of 1.0 µM

[14C]-DLM with 0.01% HSA at 37ºC for 15 min. Results are expressed as the Mean ±

SD (n = 3). (B) Effect of CSA (25 µM) on the protein binding of 1.0 µM [14C]-DLM with

4% HSA at 37ºC for 15 min. CSA (25 µM) was added simultaneously to the DLM/protein

mixture as described in the methods. Results are expressed as the Mean ± SD (n = 3).