depression gets old fast: do stress and depression accelerate cell aging?

12
Review DEPRESSION AND ANXIETY 27 : 327–338 (2010) DEPRESSION GETS OLD FAST: DO STRESS AND DEPRESSION ACCELERATE CELL AGING? Owen M. Wolkowitz, M.D., 1 Elissa S. Epel, Ph.D., 1 Victor I. Reus, M.D., 1 and Synthia H. Mellon, Ph.D. 2 Depression has been likened to a state of ‘‘accelerated aging,’’ and depressed individuals have a higher incidence of various diseases of aging, such as cardiovascular and cerebrovascular diseases, metabolic syndrome, and demen- tia. Chronic exposure to certain interlinked biochemical pathways that mediate stress-related depression may contribute to ‘‘accelerated aging,’’ cell damage, and certain comorbid medical illnesses. Biochemical mediators explored in this theoretical review include the hypothalamic–pituitary–adrenal axis (e.g., hyper- or hypoactivation of glucocorticoid receptors), neurosteroids, such as dehydro- epiandrosterone and allopregnanolone, brain-derived neurotrophic factor, excitotoxicity, oxidative and inflammatory stress, and disturbances of the telomere/telomerase maintenance system. A better appreciation of the role of these mediators in depressive illness could lead to refined models of depression, to a re-conceptualization of depression as a whole body disease rather than just a ‘‘mental illness,’’ and to the rational development of new classes of medications to treat depression and its related medical comorbidities. Depression and Anxiety 27:327–338, 2010. r r 2010 Wiley-Liss, Inc. Key words: depression; stress; aging; cortisol; BDNF; DHEA; telomeres; oxidation; inflammation; allopregnanolone Depression has been likened to a state of ‘‘accelerated aging,’’ affecting the hippocampus and the cardiovas- cular (CV), cerebrovascular, neuroendocrine, meta- bolic, and immune systems, [1–3] and depressed individuals have a higher incidence of various diseases often associated with aging, such as Type II diabetes, metabolic syndrome, osteoporosis, CV disease, stroke, and pathological cognitive aging, including Alzheimer’s disease and other dementias. [2,4–8] Depression is also associated with significantly worse outcomes in a number of medical conditions, and depression is an independent risk factor for early mortality (even after accounting for sociodemographic factors, suicide, and biological and behavioral risk factors, such as smoking, alcohol, and physical illness). [9–13] Various explanations for ‘‘accelerated aging’’ in depression have been proposed, such as the ‘‘glucocorticoid cascade’’ hypothesis [14,15] and ‘‘allostatic load.’’ [16] In this review, we explore the additional possibility that ‘‘accelerated aging’’ in depression occurs at the level of the individual cell and that it can be traced to specific biochemical mediators that are altered in depression. Discovering pathological processes in depression at the cellular level could help identify novel targets for treating depression and its comorbid medical conditions. We propose a depression model that accounts for certain linked pathogenic processes, which occur in the brain and in the periphery, and which can culminate in Published online in Wiley InterScience (www.interscience.wiley.com). DOI 10.1002/da.20686 Received for publication 9 November 2009; Revised 12 February 2010; Accepted 13 February 2010 The authors disclose the following financial relationships within the past 3 years: Contract grant sponsors: O’Shaughnessy Foundation; University of California. Correspondence to: Owen M. Wolkowitz, 401 Parnassus Ave., Box F-0984, San Francisco, CA 94143-0984. E-mail: Owen. [email protected] 1 Department of Psychiatry, University of California School of Medicine, San Francisco, California 2 Department of OB-GYN and Reproductive Sciences, University of California School of Medicine, San Francisco, California r r 2010 Wiley-Liss, Inc.

Upload: owen-m-wolkowitz

Post on 11-Jun-2016

212 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Depression gets old fast: do stress and depression accelerate cell aging?

Review

DEPRESSION AND ANXIETY 27 : 327–338 (2010)

DEPRESSION GETS OLD FAST: DO STRESS ANDDEPRESSION ACCELERATE CELL AGING?

Owen M. Wolkowitz, M.D.,1� Elissa S. Epel, Ph.D.,1 Victor I. Reus, M.D.,1 and Synthia H. Mellon, Ph.D.2

Depression has been likened to a state of ‘‘accelerated aging,’’ and depressedindividuals have a higher incidence of various diseases of aging, such ascardiovascular and cerebrovascular diseases, metabolic syndrome, and demen-tia. Chronic exposure to certain interlinked biochemical pathways that mediatestress-related depression may contribute to ‘‘accelerated aging,’’ cell damage,and certain comorbid medical illnesses. Biochemical mediators explored in thistheoretical review include the hypothalamic–pituitary–adrenal axis (e.g., hyper-or hypoactivation of glucocorticoid receptors), neurosteroids, such as dehydro-epiandrosterone and allopregnanolone, brain-derived neurotrophic factor,excitotoxicity, oxidative and inflammatory stress, and disturbances of thetelomere/telomerase maintenance system. A better appreciation of the role ofthese mediators in depressive illness could lead to refined models of depression, toa re-conceptualization of depression as a whole body disease rather than just a‘‘mental illness,’’ and to the rational development of new classes of medicationsto treat depression and its related medical comorbidities. Depression andAnxiety 27:327–338, 2010. rr 2010 Wiley-Liss, Inc.

Key words: depression; stress; aging; cortisol; BDNF; DHEA; telomeres;oxidation; inflammation; allopregnanolone

Depression has been likened to a state of ‘‘acceleratedaging,’’ affecting the hippocampus and the cardiovas-cular (CV), cerebrovascular, neuroendocrine, meta-bolic, and immune systems,[1–3] and depressedindividuals have a higher incidence of various diseasesoften associated with aging, such as Type II diabetes,metabolic syndrome, osteoporosis, CV disease, stroke,and pathological cognitive aging, including Alzheimer’sdisease and other dementias.[2,4–8] Depression is alsoassociated with significantly worse outcomes in anumber of medical conditions, and depression is anindependent risk factor for early mortality (even afteraccounting for sociodemographic factors, suicide,and biological and behavioral risk factors, such assmoking, alcohol, and physical illness).[9–13] Variousexplanations for ‘‘accelerated aging’’ in depressionhave been proposed, such as the ‘‘glucocorticoidcascade’’ hypothesis[14,15] and ‘‘allostatic load.’’[16] Inthis review, we explore the additional possibilitythat ‘‘accelerated aging’’ in depression occurs at thelevel of the individual cell and that it can be traced tospecific biochemical mediators that are altered indepression. Discovering pathological processes in

depression at the cellular level could help identifynovel targets for treating depression and its comorbidmedical conditions.

We propose a depression model that accounts forcertain linked pathogenic processes, which occur in thebrain and in the periphery, and which can culminate in

Published online in Wiley InterScience (www.interscience.wiley.com).

DOI 10.1002/da.20686

Received for publication 9 November 2009; Revised 12 February

2010; Accepted 13 February 2010

The authors disclose the following financial relationships within the past3 years: Contract grant sponsors: O’Shaughnessy Foundation;

University of California.

�Correspondence to: Owen M. Wolkowitz, 401 Parnassus Ave.,

Box F-0984, San Francisco, CA 94143-0984. E-mail: Owen.

[email protected]

1Department of Psychiatry, University of California School of

Medicine, San Francisco, California2Department of OB-GYN and Reproductive Sciences,

University of California School of Medicine, San Francisco,

California

rr 2010 Wiley-Liss, Inc.

Page 2: Depression gets old fast: do stress and depression accelerate cell aging?

cellular aging and damage and disease (Fig. 1). There iswidespread recognition that certain physical stressors,such as oxidative and inflammatory stress, can accel-erate aging in cells.[17–21] It has recently beenappreciated that psychological stress can also prema-turely age cells, possibly by invoking similar physical

processes.[15,20,22–32] Major depression and its asso-ciated biological perturbations are the focus of thisreview. To the extent similar processes are seen in otherconditions (e.g., chronic psychological stress, posttrau-matic stress disorder, schizophrenia, certain neurode-generative disorders, etc.), aspects of this model mightalso be applicable. Indeed, some of the data supportingthis model were derived from chronically stressed, butnot necessarily depressed, populations; such data willbe identified in the text. In brief, stress-relateddysregulation of the hypothalamic–pituitary–adrenal(HPA) axis, as moderated by genetic[33–36] and epige-netic[37] factors and by cognitive appraisal,[38,39] socialsupport,[40,41] and coping styles,[42,43] leads to cortisol-induced changes in gene expression (including genesrelated to monoaminergic and peptidergic neurotrans-mission), neuroendangering or neurotoxic effects incertain brain areas (e.g., prefrontal cortex and hippo-campus), excitotoxicity, oxidative stress, immunealterations leading to a proinflammatory milieu(or ‘‘neuroinflammation’’), and accelerated cell aging(via effects on the telomere/telomerase maintenancesystem), as described below. In this context, normalcompensatory or reparative processes are diminished,e.g., decreased counterregulatory neurosteroids (e.g.,dehydroepiandrosterone [DHEA]) and allopregnano-lone), decreased antioxidant compounds (e.g., VitaminC or E), diminished anti-inflammatory/immunomodu-latory cytokines (e.g., IL-10), decreased activity ofneurotrophic factors (e.g., brain-derived neurotrophicfactor [BDNF]) and decreased activity or effectivenessof the telomere-lengthening enzyme, telomerase. Thejuxtaposition of enhanced toxic processes with dimin-ished protective or restorative ones can culminate incellular damage and physical disease[44] (Table 1). Thepresentation of this model here will be relativelyconcise, but related reviews of this and similar modelsare published elsewhere.[20,22,23,36,45–53] This reviewrepresents an update and refinement of modelswe have presented earlier.[45–48] This model is notmeant to be complete or all-encompassing, nor is itmeant to apply to all individuals with major depression,because different endophenotypes of depressioncould well have different underlying pathologiccomponents.[45,54,55] Instead, this broadly sketchedmodel is meant to highlight and connect certaininteresting new findings in the study of stressand depression, and to provide testable hypothesesthat could guide research and treatment in newdirections.

GLUCOCORTICOIDS ANDNEUROSTEROIDS

The physiological significance of increased circulat-ing GC levels remains unknown, and it is evendebatable whether ‘‘hypercortisolemia’’ results in nethypercortisol-ism at the cellular level, or rather in net

)3(

(1)

)2(

)4(

)5(

)6(

)7(

)31(

)21(

GR

DHEA Allopreg CRH

Cortisol

GlucoseAvailability

Calcium; OxidativeStress

Telom Length; Telomerase

InflammatoryCytokines

BDNF

Cell Endangerment and Medical/ Psychiatric Symptoms

Glutamate; Excitotoxicity

(1, 10, 11)•••••••••••• (1, 8, 9)•••••••

Chronic Stress; Glucocorticoids

Figure 1. Theoretical model of cell ‘‘aging’’ or cell endanger-ment in depression. This overly simplified schematic model isexplained in the text (last paragraph). The mediators presentedhere are plausibly related to cell damage or dysfunction, but itremains unknown whether such cellular effects translate intopsychiatric and medical symptoms. Bracketed numbers refer topotential sites of therapeutic intervention, also described in thetext. Not depicted in this model are important monoaminergicmediators, which interact with many of the mediators that aredepicted here, as well as certain mediators not discussed in thisreview, e.g., neuropeptide Y[51,97] and gamma-aminobutyricacid.[48,212] Also not depicted here are important moderators ofstress and other effects, such as genetic polymorphisms,[33–36]

epigenetic changes,[37,67,167,168] and early life adver-sity.[52,67,213–215] To reduce the complexity of the Figure, wealso do not depict multiple linked pathways between theindividual mediators, because many of them are intertwinedand multidetermined, and we also do not indicate compensatorypathways, which can attenuate certain of the proposed effects(e.g.,[112,160]). Abbreviations: GR, glucocorticoid receptor; CRH,corticotropin releasing hormone; DHEA, dehydroepiandro-sterone; Allopreg, allopregnanolone; BDNF, brain-derivedneurotrophic factor; Telom, telomere.

328 Wolkowitz et al.

Depression and Anxiety

Page 3: Depression gets old fast: do stress and depression accelerate cell aging?

hypocortisolism, perhaps due to downregulation of theglucocorticoid receptor (GR) (referred to as ‘‘GCresistance’’).[45,56] It is possible but not proven thathyper- and hypocortisolism identify different subtypesof depression or map onto different symptom clus-ters.[45,54,57–59] It should also be recognized that theeffects of either state are likely to differ, depending onthe target tissue involved and that ‘‘relative’’ conditionsof either hyper- or hypocortisolism may exist at thesame time within organisms, making any globalstatements a simplification of the underlying endocrinestate.[60–63] For example, different GR polymorphismscan significantly affect individuals’ responses toGCs,[35,64] and alternative splicing of the GR mRNAcan lead to different GR isoforms with different actionsin different tissues.[65,66] Furthermore, early life events,such as childhood abuse, can epigenetically reprogramGR expression and splicing, leading to important inter-individual differences in GC responsivity.[67] The‘‘hypocortisolism’’ hypothesis is supported by findingsthat proinflammatory cytokine levels (e.g., tumornecrosis factor [TNF]-a, IL-1b, and IL-6) tend to beincreased in the plasma of depressed patients, and thatproinflammatory cytokines can contribute to depres-sive symptomatology. Because cortisol typically hasanti-inflammatory actions and suppresses proinflam-matory cytokines (although there are instances to thecontrary[68–71]), the coexistence of elevated cortisol andproinflammatory cytokine levels suggests an insensi-tivity to cortisol at the level of the lymphocyte GR.[72]

This possibility is supported by the finding thatperipheral GR sensitivity in depressed individuals(assessed by cutaneous vasoconstrictive responses totopically applied GCs) is inversely correlated withTNF-a concentrations.[73] The ‘‘hypocortisolism’’ hypo-thesis is also supported by recent genome-wideexpression microarray analyses on monocytes fromstressed (but not necessarily depressed) caregiverscompared to controls.[74] Despite having similarcortisol secretory patterns, the caregivers in thatstudy showed diminished expression of glucocorticoidresponse element transcripts and heightened expression

of transcripts with response elements for NF-kappaB, akey proinflammatory transcription factor.

On the other hand, the ‘‘hypercortisolism’’ hypothesisis supported by phenotypic somatic features suggestiveof cortisol excess and of increased end-organ cortisolsignaling in depression, e.g., osteoporosis, insulinresistance, Type II diabetes, a relative hypokalemicalkalosis accompanied by neutrophilia and lympho-cytosis, hypertension, metabolic syndrome and visceral/intra-abdominal adiposity (reviewed in Ref.[45]:).Further support of net GC over-activation is providedby evidence of altered expression of target genessuch as BDNF, which are believed to be undernegative regulatory control by cortisol.[75] It remainsdebatable whether hypercortisolism is causallyrelated to hippocampal atrophy often reported indepression.[32,76–82]

Pathologically elevated or diminished GC activitycould, via genomic mechanisms, alter transcription ofgenes involved in synthesis and degradation of mono-amine neurotransmitters and other substances,[83–87]

and could have neurobehavioral sequellae.[45] Chronichypercortisolemia, in particular, has been proposed bySapolsky and others,[14] to result in a biochemical‘‘cascade,’’ which can culminate in cell endangermentor cell death in certain hippocampal cells. In thesimplest description of this model, GC excess engen-ders a state of intracellular glucoprivation (insufficientintracellular glucose energy stores) in certain cells,impairing the ability of glia and other cells to clearsynaptic glutamate. The resulting excitotoxicity resultsin excessive release of calcium into the cytoplasm,which can contribute to oxidative damage, proteolysis,and cytoskeletal damage.[88–90] Unchecked, these pro-cesses can culminate in diminished cell viability or celldeath. For example, GCs can, via non-genomicmechanisms, directly modulate mitochondrial calciumand oxidation in an inverted U-shaped manner,with chronically elevated levels leading to cellulardamage.[91] In the present model, we expand uponthese earlier GC models by integrating effects onneurotrophic factors, neurosteroids, inflammation, and

TABLE 1. Possible detrimental changes seen in depression and/or chronic stress

mPotentially damaging mediators kPotentially protective or restorative mediators

INCREASED DECREASEDHypercortisolemia (with hyper- or hypocortisolism) Neurosteroids (allopregnanolone, DHEAa)Synaptic glutamate (excitotoxicity) Insulin sensitivityIntracytoplasmic calcium Intracellular glucoseFree radicals (oxidative stress) AntioxidantsInflammatory cytokines Anti-inflammatory/immunoregulatory cytokinesb

Neurotrophic factorsTelomerasec

aEvidence is mixed as to whether major depression is characterized by excessive or diminished levels of DHEA, but it is often low withpsychological stress.bEvidence is mixed as to whether the anti-inflammatory/immunoregulatory cytokine, IL-10, is elevated or diminished in major depression.cTelomerase activity has been reported as low or high (albeit less effective in preserving telomere length) in chronic stress; there are as yet nopublished data on telomerase activity in major depression.

329Review: Do Stress and Depression Accelerate Cell Aging?

Depression and Anxiety

Page 4: Depression gets old fast: do stress and depression accelerate cell aging?

the telomere/telomerase maintenance system, an im-portant aspect of cell aging.

Although circulating cortisol concentrations arefrequently elevated in depression, plasma and CSFconcentrations of the GABA-A receptor agonistneurosteroid, allopregnanolone, are decreased in unme-dicated depressives, plasma and CSF levels of allo-pregnanolone increase with selective serotoninreuptake inhibitor (SSRI) treatment in proportion totheir antidepressant effect.[92,93] SSRI antidepressantsrapidly increase allopregnanolone synthesis, and thismay contribute to their anxiolytic effects.[92,94,95]

Another neurosteroid, DHEA, which can have ‘‘anti-cortisol’’ effects (reviewed in[96]), and which promotespsychological resilience,[51,97] has been reported to beboth high and low in depression,[96] but DHEAtreatment is generally reported as having significantantidepressant effects.[96] Notably, both theseneurosteroids modulate HPA,[96] BDNF[96,98,99] andimmune[3,100–102] activity, antagonize oxidativestress[103,104] and have neuroprotective or neuroregen-erative effects.[96,98–101] Allopregnanolone also inhbitsstress-induced corticotropin-releasing hormone re-lease.[99] Endogenous decreases in these neurosteroidsor exogenously produced increases in their effectswould be expected to have damaging or beneficialeffects, respectively, in the context of depression orchronic stress.[48,92,95,96,105,106]

IMMUNE FUNCTIONStress-related dysregulation of HPA axis and of GC

activity also contributes to immune dysregulation indepression,[107] and proinflammatory cytokines furtheralter HPA axis activity.[108,109] Immune dysregulationmay be an important pathway by which depression andchronic stress heighten the risk of serious medicalcomorbidity.[20,30,102,110,111] Several major proinflam-matory cytokines, such as IL-1b, IL-2, IL-6, andTNF-a, are elevated in depression, either basally or inresponse to mitogen stimulation or acutestress.[107,112,113] Conversely, certain anti-inflammatoryor immunomodulatory cytokines, such as IL-1 receptorantagonist and IL-10, may be increased or decreased ormay be dysregulated relative to proinflammatorycytokines.[112,114,115] In particular, the ratio of proin-flammatory to anti-inflammatory/immunomodulatorycytokines may be heightened in depression and couldresult in increased inflammation[112] and, subsequently,in increased free radical production and oxidativestress.[116] Converging findings suggest that highperipheral levels of inflammatory cytokines, such asIL-6, are associated with the activation of centralinflammatory mechanisms that, under some circum-stances, adversely affect the hippocampus, where IL-6receptors are abundantly expressed.[117] Hippocampalneurogenesis is also suppressed by microglial acti-vation, which leads to brain inflammation,[118] andhigh proinflammatory cytokine concentrations can

contribute to hippocampal neurodegeneration.[119] Inwild-type mice, stress increases hippocampal IL-6concentrations, but IL-6 (�/�) knockout mice areresistant to stress-induced learned helplessness, ananimal model of depression.[120] In healthy humans,plasma IL-6 concentrations are inversely correlatedwith hippocampal gray matter,[121] and elevated pre-treatment inflammatory cytokine levels predict poorerresponse to antidepressant medications in individualswith major depression.[122] High proinflammatorycytokine levels also directly contribute to monoaminedysregulation, HPA axis stimulation, depression, andcellular and organismic senescence.[119,123] It should benoted, however, that due to the complexity of cytokineactions in neurons and glia, the end effect of individualcytokines can be either detrimental or protective,depending on the circumstances.[112]

OXIDATIONStress and altered HPA axis activity can also

increase oxidative damage and decrease antioxidantdefenses.[20,29,46,124] Oxidative stress, together withinflammatory cytokines, often increase with aging andin various disease states, whereas antioxidant and anti-inflammatory activities paradoxically decrease, result-ing in a heightened likelihood of cellular damage and ofa senescent phenotype.[20,125] Oxidative stress occurswhen the production of oxygen-free radicals exceedsthe capacity of the body’s antioxidants to neutralizethem. Oxidative stress damages DNA, protein, lipids,and other macromolecules in many tissues, withtelomeres (discussed below)[126] and the brain[90] beingparticularly sensitive. Elevated plasma and/or urineoxidative stress markers (e.g., increased F2-isopros-tanes and 8-hydroxydeoxyguanosine [8-OHdG] alongwith decreased antioxidant compounds, such as Vita-mins C and E) have been reported in individuals withdepression and in those with chronic psychologicalstress,[27,29,127,128] and the concentration of peripheraloxidative stress markers is positively correlated with theseverity and chronicity of depression,[29,129,130] as wellas with evidence of accelerated apoptosis in polymor-phonuclear blood cells.[131] Furthermore, the ratio ofserum oxidized lipids (F2-isoprostanes) to antioxidants(Vitamin E) is directly related to psychological stress,and is inversely related to telomere length andtelomerase activity (both discussed below) in chroni-cally stressed caregivers.[22] Conversely, antidepressantsdecrease oxidative stress.[132] Because cellular oxidativedamage is an important component of the aging process,prolonged or repeated exposure to oxidative stress couldaccelerate aspects of biological aging and promoteaging-related comorbid diseases in depression.[29] Forexample, oxidative stress potentiates TNF-a-inducedactivation of the cell death cascade.[133] Stress- ordepression-related increases in oxidative stress addition-ally blunt certain protective or reparative processes,because oxidative stress is inversely correlated with

330 Wolkowitz et al.

Depression and Anxiety

Page 5: Depression gets old fast: do stress and depression accelerate cell aging?

telomerase activity as well as telomere length (discussedbelow),[126,134] and because increased oxidative stress(and lower antioxidant protection) is associated withlower BDNF activity[135,136] (discussed below).

BRAIN-DERIVEDNEUROTROPHIC FACTOR

The ‘‘neurotrophic model’’ of depression[75] posits thatdiminished hippocampal BDNF activity, caused by stressor excessive GCs, impairs the ability of stem cells in thesubgranular zone of the dentate gyrus (as well as cells inthe subventricular zone, projecting to the prefrontalcortex) to proliferate into mature cells that remain viable.It is not known whether such processes can causedepression and whether they are relevant to the mechan-ism of action of antidepressant drugs; evidence issomewhat stronger for BDNF involvement in antide-pressant effects than in the etiology of depression.[137,138]

Furthermore, unmedicated patients with depression havedecreased hippocampal (at autopsy) and serum concen-trations of BDNF.[137,139,140] A role of BDNF inantidepressant mechanisms of action is supported byfindings that hippocampal neurogenesis (in animals) andserum BDNF concentrations (in depressed humans)increase with antidepressant treatment,[137,140] and thathippocampal neurogenesis is required for behavioraleffects of antidepressants in animals.[141] Apart from itsdirect neurotrophic actions, BDNF also has anti-inflammatory and antioxidant effects and improves theefficiency of brain mitochondrial oxygen utilization,which may contribute to its neuroprotective effi-cacy,[142,143] BDNF attenuates glucocorticoid-inducedneuronal death,[144] and BDNF activity synergizes withtelomerase activity (discussed below) in promoting thegrowth of developing neurons.[145]

CELL AGING: TELOMERES ANDTELOMERASE

Telomeres are DNA-protein complexes that cap theends of linear DNA strands, protecting DNA fromdamage.[146] When telomeres reach a critically shortlength, as happens when cells undergo repeated mitoticdivisions without adequate telomerase activity (e.g.,immune cells and stem cells, including neurogenic stemcells in the hippocampus), cells become susceptible toapoptosis and death. Even in nondividing cells, such asmature neurons, telomeres can become shortened byoxidative stress, which preferentially damages telo-meres to a greater extent than nontelomericDNA.[126,147] This non-mitotic type of telomere short-ening also increases susceptibly to apoptosis and celldeath. Telomere length is a indicator of ‘‘biologicalage’’ (as opposed to just chronological age) andrepresents a cumulative log of the number of celldivisions and a cumulative record of exposure togenotoxic and cytotoxic processes, such as oxida-

tion.[20,22,23,126,146,148] Telomere length may also repre-sent a biomarker for assessing an individual’scumulative exposure to, or ability to cope with, stressfulconditions. For example, preliminary data point toaccelerated leukocyte telomere shortening, a sign ofcellular aging, in chronically stressed[22,23] and indepressed[149] individuals. The telomere shorteningmay, at least in part, be related to increases in stress-related cortisol and catecholamine output.[23,150] Theestimated magnitude of the acceleration of biologicalaging is not trivial; it was estimated as approximately9–17 additional years of chronological aging in thestressed caregivers and as much as 10 years in thedepressed individuals. It should be noted that thesubjects in the depression study had very chroniccourses of depression (an average of nearly 26 years oflifetime depression).[149] Preliminary data suggest thattelomere shortening is a function of the duration of thelifetime exposure to depression (Wolkowitz et al.,unpublished) and may not be present in individualswith short lifetime exposures to depression. In non-depressed populations, shortening of leukocyte telo-meres is associated with atherosclerosis and CVdisease,[151–153] osteoporosis[154] and cognitive impair-ment,[155] and with increased medical morbidity andearlier mortality from a number of causes, includingCV and infectious disease, and dementia.[156] Forexample, shortened telomeres are associated with agreater than three-fold increase in the risk of myocar-dial infarction and stroke, and with a greater than eight-fold increase in the risk of death from infectiousdisease.[157] In a more recent study, baseline telomerelength (in women) and prospective rate of change intelomere length over a 2.5 year period (in men)predicted CV mortality over a 12-year period.[156]

Thus, cell aging (manifest as shortened telomeres),associated with any of the mediators discussed above,provides a conceptual link between depression and itsassociated medical comorbidities and shortenedlife span.[20,102,148]

Telomerase is a reverse transcriptase enzyme thatrebuilds telomere length, thereby delaying cell senes-cence, apoptosis, and cell death.[146] Telomerase alsohas antiaging or cell survival-promoting effects in-dependent of its effects on telomere length byregulating transcription of growth factors, synergizingwith the neurotrophic effects of BDNF, havingantioxidant effects and intrinsic antiapoptotic effects,protecting cells from necrosis, and stimulating cellgrowth in adverse conditions.[145,158,159] Telomeraseactivity has not yet been characterized in individualswith major depression, but it has been reported to bediminished[22] or increased[160] in stressed caregiverscompared to low stress controls. Several of themediators discussed above can contribute to dimin-ished telomere length and/or telomerase activity(e.g., cortisol,[150] oxidative stress,[147] and inflamma-tory cytokines[160,161]), highlighting the interlinkednature of cell-damaging and cell-protective mediators

331Review: Do Stress and Depression Accelerate Cell Aging?

Depression and Anxiety

Page 6: Depression gets old fast: do stress and depression accelerate cell aging?

in stress and depression. Important moderators oftelomere length are rapidly being discovered (e.g.,childhood maltreatment,[162] socioeconomic status,[163]

race,[164,165] physical exercise,[166] and dispositionalpessimism,[39] among others).

DO STRESS AND DEPRESSIONACCELERATE CELL AGING?

We have briefly reviewed evidence of biochemicalabnormalities in depression, some of which areconsistent with an aged phenotype that could con-tribute to certain medical comorbidities seen withdepression. They could also contribute to the depres-sive state itself, but that has not been adequately tested.In particular, depression (and perhaps chronic stress, aswell) may be associated with increased cell damagingprocesses and decreased cell protective or restorativeones (Table 1). We propose a model in which theseabnormalities are causally interlinked and may derive,directly or indirectly, from altered HPA axis and GCactivity seen in depression (Fig. 1). It remains uncertainwhether the brain in depressed individuals is subject tonet hypo- or hypercortisolism, and even within thebrain, individual component tissues, such as neuronsand glia, may differ in their response to alteredcirculating GC levels as a result of differing receptorexpression or metabolic enzymes.

We have couched this model in terms of ‘‘acceleratedaging’’ at the cell level, although whether cell aging isactually accelerated in depression remains to bedetermined in prospective trials. It is important torecognize that this model is unlikely to apply to allindividuals with depression (many of whom do nothave discernible HPA axis dysregulation), and thatmany of these changes are not specific to majordepression. Also, various genetic and epigenetic mod-erators, not discussed here, are undoubtedly impor-tant.[39,52,67,167–169] The major importance of thishypothetical model is that it identifies certain non-traditional targets for pharmacological and nonphar-macological treatment, and thus could lead to newtheory-driven therapies. In particular, treatments di-rected at the targets identified here have the potentialnot only to treat depression but also to treatcertain medical comorbidities that occur alongsidedepression.[47,49,170] Interestingly, even traditionalantidepressant medications, which putatively workvia monoaminergic actions, affect many of the noveltargets described here[95,109,128,171–177] (see Fig. 1), eventhough they were not developed with those purposes inmind. Last, the identification of novel biomarkers ofdepression may discriminate separate endophenotypesof depression that respond differently to differenttreatments,[54,55,122,174] although some of the endocri-nological and neurochemical differences reported maybe dependent more on the target tissue examined thanreflective of a global endophenotype. This will hope-

fully accelerate the era of personalized antidepressanttreatment.

THEORETICAL MODEL ANDNOVEL TREATMENT

POSSIBILITIESA schematic overview of our model is presented in

Figure 1. The condensed and simplified nature of thisschematic precludes depiction of numerous othermediators and moderators and interactions that areinvolved. Therefore, this depiction should be viewedas a ‘‘broad brush stroke’’ theoretical model. Thebracketed numbers in Figure 1 are keyed to potentialsites of therapeutic intervention described below. Inthis model, elevated cortisol levels are associated withdownregulation of GRs (‘‘GC resistance’’); the ‘‘net’’GC activity remains uncertain and could even differ indifferent tissues. A deficit in GR function can precedeor result from the hypercortisolemia. To the extent thatlymphocyte GRs become GC resistant, immunefunction is altered and excessive proinflammatorycytokine effects can occur. Changes in cortisol activityalso result in multiple genomic changes, e.g., alteredlevels of certain neurotransmitters (e.g., decreasedserotonin and increased dopamine activity in certainbrain regions, which could contribute to depressive orpsychotic symptoms). To the extent GC activity is‘‘excessive’’ in certain brain regions, a cascade ofevents can follow, characterized by diminished insulinsignaling, intraneuronal glucoprivation and diminishedenergy availability, defective clearance of intrasynapticglutamate, excitotoxicity, intracellular buildup ofcalcium, generation of oxygen-free radicals (oxidativestress), diminution of telomerase activity and cellulardamage or cell death. Increased oxidative stress candamage the enzyme telomerase and shorten telomeres,at least in certain cells in the body. In nondepressedindividuals, leukocyte telomere shortening is associatedwith a host of physical illnesses and prematuremortality. If this occurs in depressed individuals aswell, it could help explain the surfeit of medical illnessand the shortened life expectancy seen with chronicdepression. Chronic stress and depression and/orexcessive cortisol exposure can also be associated withunderproduction of certain counterregulatory neuro-steroid hormones, e.g., DHEA and allopregnanolone,which could further dysregulate HPA axis activity,hamper antioxidative function, and reduce neuropro-tective capacity. Additionally, prolonged stress and/orincreased cortisol activity can downregulate BDNFactivity, which further diminishes neuroreparativecapacity and attenuates neurogenesis.

To the extent this theoretical model is accurate,several potential treatment loci emerge, as indicatednumerically in Figure 1: (1) traditional antidepressantshave several novel functions apart from increasing intra-synaptic monoamine concentrations: they up-regulate

332 Wolkowitz et al.

Depression and Anxiety

Page 7: Depression gets old fast: do stress and depression accelerate cell aging?

GR function,[172] increase allopregnanolone synthesis(certain SSRIs),[95] increase BDNF levels,[75] and haveanti-inflammatory[174,178] and antioxidant[179,180] effects;(2) CRH antagonists;[181] (3) stress reduction, medita-tion, and other behavioral and lifestyle interven-tions;[20,182,183] (4) antiglucocorticoids[47,184–186]; (5)energy supplementation or insulin receptor sensiti-zers;[187–189] (6) glutamate antagonists;[190–194] (7) cal-cium blockers[195,196] and antioxidants[197]; (8) DHEA;[96]

(9) 3-a-hydroxy–steroid dehydrogenase (3-a-HSD)stimulators (including SSRIs), which increase allopreg-nanolone synthesis[94,95]; (10) environmental enrichment,exercise[198–201]; (11) BDNF administration via novelroutes of administration[202–204]; (12) telomerase activa-tion,[205,206] and (13) anti-inflammatory drugs, TNF-aantagonists, etc.[109,174,207–211] It is possible that, bytargeting such ‘‘upstream’’ mediators of the biochemicalmilieu, additional therapeutic leverage might be gained.Already, preliminary studies are testing many of thesestrategies, with preliminary signs of success.

Acknowledgments. The authors acknowledge thegenerosity of the O’Shaughnessy Foundation, whichsupplied major funding. Additional funding wassupplied by the University of California, San Francisco,Academic Senate. The authors are also grateful toDr. Jue Lin, who has provided expert advice andtechnical aid in the field of cell aging and Dr. ElizabethBlackburn, a pioneer in the field of cell aging, whoseguidance has been indispensible.

Financial disclosures: Dr. Wolkowitz has receivedlecture honoraria from Jazz Pharmaceuticals andMerck Pharmaceuticals, and has served on an AdvisoryBoard for Pfizer Pharmaceuticals. No other authorshave financial ties to these or any other pharmaceuticalcompanies.

REFERENCES1. Heuser I. Depression, endocrinologically a syndrome of

premature aging? Maturitas 2002;41:S19–S23.2. McIntyre RS, Soczynska JK, Konarski JZ et al. Should depressive

syndromes be reclassified as ‘‘metabolic syndrome type II’’? AnnClin Psychiatry 2007;19:257–264.

3. Bauer ME. Chronic stress and immunosenescence: a review.Neuroimmunomodulation 2008;15:241–250.

4. Evans DL, Charney DS, Lewis L et al. Mood disorders in themedically ill: scientific review and recommendations. BiolPsychiatry 2005;58:175–189.

5. McIntyre RS, Rasgon NL, Kemp DE et al. Metabolic syndromeand major depressive disorder: co-occurrence and pathophysio-logic overlap. Curr Diab Rep 2009;9:51–59.

6. Brown ES, Varghese FP, McEwen BS. Association of depressionwith medical illness: does cortisol play a role? Biol Psychiatry2004;55:1–9.

7. Vogelzangs N, Suthers K, Ferrucci L et al. Hypercortisolemicdepression is associated with the metabolic syndrome in late-life.Psychoneuroendocrinology 2007;32:151–159.

8. Speck CE, Kukull WA, Brenner DE et al. History of depressionas a risk factor for Alzheimer’s disease. Epidemiology 1995;6:366–369.

9. Chodosh J, Kado DM, Seeman TE, Karlamangla AS. Depressivesymptoms as a predictor of cognitive decline: MacArthur studiesof successful aging. Am J Geriatr Psychiatry 2007;15:406–415.

10. Gump BB, Matthews KA, Eberly LE, Chang YF. Depressivesymptoms and mortality in men: results from the multiple riskfactor intervention trial. Stroke 2005;36:98–102.

11. McCusker J, Cole M, Ciampi A, Latimer E, Windholz S,Belzile E. Major depression in older medical inpatients predictspoor physical and mental health status over 12 months. GenHosp Psychiatry 2007;29:340–348.

12. Musselman DL, Evans DL, Nemeroff CB. The relationship ofdepression to cardiovascular disease: epidemiology, biology, andtreatment. Arch Gen Psychiatry 1998;55:580–592.

13. Schulz R, Beach SR, Ives DG, Martire LM, Ariyo AA, Kop WJ.Association between depression and mortality in older adults: theCardiovascular Health Study. Arch Intern Med 2000;160:1761–1768.

14. Sapolsky RM, Krey LC, McEwen BS. The neuroendocrinologyof stress and aging: the glucocorticoid cascade hypothesis.Endocr Rev 1986;7:284–301.

15. Sapolsky RM. Glucocorticoids, stress, and their adverse neuro-logical effects: relevance to aging. Exp Gerontol 1999;34:721–732.

16. McEwen BS. Protective and damaging effects of stress mediators:the good and bad sides of the response to stress. Metabolism2002;51:2–4.

17. Mattson MP, Maudsley S, Martin B. A neural signalingtriumvirate that influences ageing and age-related disease:insulin/IGF-1, BDNF and serotonin. Ageing Res Rev 2004;3:445–464.

18. McEwen JE, Zimniak P, Mehta JL, Reis RJ. Molecular pathologyof aging and its implications for senescent coronary athero-sclerosis. Curr Opin Cardiol 2005;20:399–406.

19. Voss P, Siems W. Clinical oxidation parameters of aging. FreeRadic Res 2006;40:1339–1349.

20. Epel ES. Psychological and metabolic stress: a recipe foraccelerated cellular aging? Hormones (Athens) 2009;8:7–22.

21. Sarkar D, Fisher PB. Molecular mechanisms of aging-associatedinflammation. Cancer Lett 2006;236:13–23.

22. Epel ES, Blackburn EH, Lin J et al. Accelerated telomereshortening in response to life stress. Proc Natl Acad Sci USA2004;101:17312–17315.

23. Epel ES, Lin J, Wilhelm FH et al. Cell aging in relation to stressarousal and cardiovascular disease risk factors. Psychoneuroen-docrinology 2006;31:277–287.

24. Epel ES, Lin J, Dhabhar FS et al. Dynamics of telomeraseactivity in response to acute psychological stress. Brain BehavImmun 2010, in press.

25. Sapolsky RM. Organismal stress and telomeric aging: anunexpected connection. Proc Natl Acad Sci USA 2004;101:17323–17324.

26. Forlenza MJ, Latimer JJ, Baum A. The effects of stress on DNArepair capacity. Psychol Health 2000;15:881–891.

27. Forlenza MJ, Miller GE. Increased serum levels of 8-hydroxy-2’-deoxyguanosine in clinical depression. Psychosom Med 2006;68:1–7.

28. Irie M, Asami S, Nagata S, Ikeda M, Miyata M, Kasai H.Psychosocial factors as a potential trigger of oxidative DNAdamage in human leukocytes. Jpn J Cancer Res 2001;92:367–376.

29. Irie M, Miyata M, Kasai H. Depression and possible cancer riskdue to oxidative DNA damage. J Psychiatr Res 2005;39:553–560.

333Review: Do Stress and Depression Accelerate Cell Aging?

Depression and Anxiety

Page 8: Depression gets old fast: do stress and depression accelerate cell aging?

30. Kiecolt-Glaser JK, Preacher KJ, MacCallum RC, Atkinson C,Malarkey WB, Glaser R. Chronic stress and age-related increasesin the proinflammatory cytokine IL-6. Proc Natl Acad Sci USA2003;100:9090–9095.

31. Miller DB, O’Callaghan JP. Effects of aging and stress onhippocampal structure and function. Metabolism 2003;52:17–21.

32. McEwen BS. Protective and damaging effects of stress mediators:central role of the brain. Dialogues Clin Neurosci 2006;8:367–381.

33. Wust S, Federenko IS, van Rossum EF et al. A psychobiologicalperspective on genetic determinants of hypothalamus-pituitary-adrenal axis activity. Ann N Y Acad Sci 2004;1032:52–62.

34. Kaufman J, Yang BZ, Douglas-Palumberi H et al. Brain-derivedneurotrophic factor-5-HTTLPR gene interactions and environ-mental modifiers of depression in children. Biol Psychiatry 2006;59:673–680.

35. DeRijk R, de Kloet ER. Corticosteroid receptor geneticpolymorphisms and stress responsivity. Endocrine 2005;28:263–270.

36. Charney DS, Manji HK. Life stress, genes, and depression:multiple pathways lead to increased risk and new opportunitiesfor intervention. Sci STKE 2004;2004:re5.

37. Mesquita AR, Wegerich Y, Patchev AV et al. Glucocorticoids andneuro- and behavioural development. Semin Fetal Neonatal Med2009;14:130–135.

38. Folkman S, Lazarus RS. Stress-processes and depressive symp-tomatology. J Abnorm Psychol 1986;95:107–113.

39. O’Donovan A, Lin J, Dhabhar FS et al. Pessimism correlateswith leukocyte telomere shortness and elevated interleukin-6 inpost-menopausal women. Brain Behav Immun 2009;23:446–449.

40. Ozbay F, Fitterling H, Charney D, Southwick S. Social supportand resilience to stress across the life span: a neurobiologicframework. Curr Psychiatry Rep 2008;10:304–310.

41. DeLongis A, Folkman S, Lazarus RS. The impact of daily stresson health and mood: psychological and social resources asmediators. J Pers Soc Psychol 1988;54:486–495.

42. Aldwin CM, Revenson TA. Does coping help? A reexaminationof the relation between coping and mental health. J Pers SocPsychol 1987;53:337–348.

43. Kemeny ME, Gruenewald TL. Psychoneuroimmunology update.Semin Gastrointest Dis 1999;10:20–29.

44. Mattson MP, Duan W, Chan SL et al. Neuroprotective andneurorestorative signal transduction mechanisms in brain aging:modification by genes, diet and behavior. Neurobiol Aging2002;23:695–705.

45. Wolkowitz OM, Burke H, Epel ES, Reus VI. Glucocorticoids:mood, memory and mechanisms. Ann NY Acad Sci 2009;1179:19–40.

46. Wolkowitz OM, Epel ES, Mellon S. When blue turns to grey: dostress and depression accelerate cell aging? World J BiolPsychiatry 2008;9:2–5.

47. Wolkowitz OM, Epel ES, Reus VI. Stress hormone-relatedpsychopathology: pathophysiological and treatment implications.World J Biol Psychiatry 2001;2:115–143.

48. Wolkowitz OM, Reus VI. Neurotransmitters, neurosteroids andneurotrophins: new models of the pathophysiology and treat-ment of depression. World J Biol Psychiatry 2003;4:98–102.

49. Wolkowitz OM, Epel ES, Reus VI. Antiglucocorticoid strategiesin treating major depression and improving health outcome. In:Thakore J, ed. The Physical Consequences of Depression.Petersfield, UK: Wrightson Biomedical Publishing, Ltd.;2001:181–213.

50. Epel ES, Burke H, Wolkowitz OM. The psychoneuroendocri-nology of aging. In: Aldwin CM, Park CL, Spior III A, eds.

Handbook of Health Psychology and Aging. New York: GuilfordPublications, Inc.; 2007:119–141.

51. Charney DS. Psychobiological mechanisms of resilience andvulnerability: implications for successful adaptation to extremestress. Am J Psychiatry 2004;161:195–216.

52. aan het Rot M, Mathew SJ, Charney DS. Neurobiologicalmechanisms in major depressive disorder. Can Med Assoc J 2009;180:305–313.

53. Manji HK, Duman RS. Impairments of neuroplasticity andcellular resilience in severe mood disorders: implications for thedevelopment of novel therapeutics. Psychopharmacol Bull 2001;35:5–49.

54. Hasler G, Drevets WC, Manji HK, Charney DS. Discoveringendophenotypes for major depression. Neuropsychopharmacol-ogy 2004;29:1765–1781.

55. Gottesman, II, Gould TD. The endophenotype concept inpsychiatry: etymology and strategic intentions. Am J Psychiatry2003;160:636–645.

56. Raison CL, Miller AH. When not enough is too much: the roleof insufficient glucocorticoid signaling in the pathophysiology ofstress-related disorders. Am J Psychiatry 2003;160:1554–1565.

57. Bremmer MA, Deeg DJ, Beekman AT, Penninx BW, Lips P,Hoogendijk WJ. Major depression in late life is associated withboth hypo- and hypercortisolemia. Biol Psychiatry 2007;62:479–486.

58. Fries E, Hesse J, Hellhammer J, Hellhammer DH. A new view onhypocortisolism. Psychoneuroendocrinology 2005;30:1010–1016.

59. Heim C, Ehlert U, Hellhammer DH. The potential role ofhypocortisolism in the pathophysiology of stress-related bodilydisorders. Psychoneuroendocrinology 2000;25:1–35.

60. Ebrecht M, Buske-Kirschbaum A, Hellhammer DSK, RohlederNBW, Kirschbaum C. Tissue specificity of glucocorticoid sensiti-vity in healthy adults. J Clin Endocrinol Metab 2000;85:3733–3739.

61. Webster MJ, Knable MB, O’Grady J, Orthmann J, Weickert CS.Regional specificity of brain glucocorticoid receptor mRNAalterations in subjects with schizophrenia and mood disorders.Mol Psychiatry 2002;7:985–994.

62. Lu NZ, Cidlowski JA. Glucocorticoid receptor isoforms generatetranscription specificity. Trends Cell Biol 2006;16:301–307.

63. Yudt MR, Cidlowski JA. The glucocorticoid receptor: coding adiversity of proteins and responses through a single gene. MolEndocrinol 2002;16:1719–1726.

64. Russcher H, Smit P, van den Akker EL et al. Two polymorphismsin the glucocorticoid receptor gene directly affect glucocorti-coid-regulated gene expression. J Clin Endocrinol Metab 2005;90:5804–5810.

65. Zhou J, Cidlowski JA. The human glucocorticoid receptor: onegene, multiple proteins and diverse responses. Steroids 2005;70:407–417.

66. Chrousos GP, Kino T. Glucocorticoid action networks and complexpsychiatric and/or somatic disorders. Stress 2007;10:213–219.

67. McGowan PO, Sasaki A, D’Alessio AC et al. Epigenetic regula-tion of the glucocorticoid receptor in human brain associateswith childhood abuse. Nat Neurosci 2009;12:342–348.

68. Frank MG, Miguel ZD, Watkins LR, Maier SF. Prior exposureto glucocorticoids sensitizes the neuroinflammatory and peri-pheral inflammatory responses to E. coli lipopolysaccharide.Brain Behav Immun 2010;24:19–30.

69. MacPherson A, Dinkel K, Sapolsky R. Glucocorticoids worsenexcitotoxin-induced expression of pro-inflammatory cytokines inhippocampal cultures. Exp Neurol 2005;194:376–383.

70. Sorrells SF, Sapolsky RM. A pro-inflammatory review ofglucocorticoid actions in the CNS. Brain Behav Immun 2007;21:259–272.

334 Wolkowitz et al.

Depression and Anxiety

Page 9: Depression gets old fast: do stress and depression accelerate cell aging?

71. Schuld A, Schmid DA, Haack M, Holsboer F, Friess E,Pollmacher T. Hypothalamo-pituitary-adrenal function inpatients with depressive disorders is correlated with baselinecytokine levels, but not with cytokine responses to hydrocorti-sone. J Psychiatr Res 2003;37:463–470.

72. Pace TW, Hu F, Miller AH. Cytokine-effects on glucocorticoidreceptor function: relevance to glucocorticoid resistance and thepathophysiology and treatment of major depression. Brain BehavImmun 2007;21:9–19.

73. Fitzgerald P, O’Brien SM, Scully P, Rijkers K, Scott LV,Dinan TG. Cutaneous glucocorticoid receptor sensitivity andpro-inflammatory cytokine levels in antidepressant-resistantdepression. Psychol Med 2006;36:37–43.

74. Miller GE, Chen E, Sze J et al. A functional genomic fingerprintof chronic stress in humans: blunted glucocorticoid and increasedNF-kappaB signaling. Biol Psychiatry 2008;64:266–272.

75. Duman RS, Monteggia LM. A neurotrophic model for stress-related mood disorders. Biol Psychiatry 2006;59:1116–1127.

76. Muller MB, Lucassen PJ, Yassouridis A, Hoogendijk WJ,Holsboer F, Swaab DF. Neither major depression nor gluco-corticoid treatment affects the cellular integrity of the humanhippocampus. Eur J Neurosci 2001;14:1603–1612.

77. Lucassen PJ, Heine VM, Muller MB et al Stress, depression andhippocampal apoptosis. CNS Neurol Disord Drug Targets2006;5:531–546.

78. Campbell S, Marriott M, Nahmias C, MacQueen GM. Lowerhippocampal volume in patients suffering from depression: ameta-analysis. Am J Psychiatry 2004;161:598–607.

79. Videbech P, Ravnkilde B. Hippocampal volume and depression: ameta-analysis of MRI studies. Am J Psychiatry 2004;161:1957–1966.

80. Sheline YI. 3D MRI studies of neuroanatomic changes inunipolar major depression: the role of stress and medical co-morbidity. Biol Psychiatry 2000;48:791–800.

81. Bremner JD. Does stress damage the brain? Biol Psychiatry1999;45:797–805.

82. Sapolsky RM. Glucocorticoids and hippocampal atrophy inneuropsychiatric disorders. Arch Gen Psychiatry 2000;57:925–935.

83. Dinan TG. Noradrenergic and serotonergic abnormalities indepression: stress- induced dysfunction? J Clin Psychiatry 1996;57:14–18.

84. McEwen BS. Glucocorticoid-biogenic amine interactions inrelation to mood and behavior. Biochem Pharmacol 1987;36:1755–1763.

85. Wolkowitz OM, Doran AR, Breier A et al. The effects ofdexamethasone on plasma homovanillic acid and 3-methoxy-4-hydroxyphenylglycol. Evidence for abnormal corticosteroid-catecholamine interactions in major depression. Arch GenPsychiatry 1987;44:782–789.

86. Wolkowitz OM. Prospective controlled studies of the behavioraland biological effects of exogenous corticosteroids. Psychoneuro-endocrinology 1994;19:233–255.

87. Lesch KP, Lerer B. The 5-HT receptor—G-protein—effectorsystem complex in depression. I. Effect of glucocorticoids.J Neural Transm Gen Sect 1991;84:3–18.

88. Lee AL, Ogle WO, Sapolsky RM. Stress and depression: possiblelinks to neuron death in the hippocampus. Bipolar Disord2002;4:117–128.

89. Sapolsky RM. The possibility of neurotoxicity in the hippo-campus in major depression: a primer on neuron death. BiolPsychiatry 2000;48:755–765.

90. Facheris M, Beretta S, Ferrarese C. Peripheral markers ofoxidative stress and excitotoxicity in neurodegenerative disorders:

tools for diagnosis and therapy? J Alzheimers Dis 2004;6:177–184.

91. Du J, McEwen B, Manji HK. Glucocorticoid receptors modulatemitochondrial function: a novel mechanism for neuroprotection.Commun Integr Biol 2009;2:350–352.

92. Uzunova V, Sheline Y, Davis JM et al. Increase in thecerebrospinal fluid content of neurosteroids in patients withunipolar major depression who are receiving fluoxetine orfluvoxamine. Proc Natl Acad Sci U S A 1998;95:3239–3244.

93. Strohle A, Romeo E, Hermann B et al. Concentrations of3a-reduced neuroactive steroids and their precursors in plasma ofpatients with major depression and after clinical recovery. BiolPsychiatry 1999;45:274–277.

94. Griffin LD, Mellon SH. Selective serotonin reuptake inhibitorsdirectly alter activity of neurosteroidogenic enzymes. Proc NatlAcad Sci 1999;96:13512–13517.

95. Guidotti A, Costa E. Can the antidysphoric and anxiolyticprofiles of selective serotonin reuptake inhibitors be related totheir ability to increase brain allopregnanolone availability? BiolPsychiatry 1998;44:865–873.

96. Maninger N, Wolkowitz OM, Reus VI, Epel ES, Mellon SH.Neurobiological and neuropsychiatric effects of dehydroepian-drosterone (DHEA) and DHEA sulfate (DHEAS). FrontNeuroendocrinol 2009;30:65–91.

97. Yehuda R, Flory JD, Southwick S, Charney DS. Developing anagenda for translational studies of resilience and vulnerabilityfollowing trauma exposure. Ann N Y Acad Sci 2006;1071:379–396.

98. Naert G, Maurice T, Tapia-Arancibia L, Givalois L. Neuroactivesteroids modulate HPA axis activity and cerebral brain-derivedneurotrophic factor (BDNF) protein levels in adult male rats.Psychoneuroendocrinology 2007;32:1062–1078.

99. Patchev VK, Hassan AH, Holsboer DF, Almeida OF. Theneurosteroid tetrahydroprogesterone attenuates the endocrineresponse to stress and exerts glucocorticoid-like effects onvasopressin gene transcription in the rat hypothalamus. Neuro-psychopharmacology 1996;15:533–540.

100. Wang JM, Irwin RW, Liu L, Chen S, Brinton RD. Regenera-tion in a degenerating brain: potential of allopregnanoloneas a neuroregenerative agent. Curr Alzheimer Res 2007;4:510–517.

101. He J, Evans CO, Hoffman SW, Oyesiku NM, Stein DG.Progesterone and allopregnanolone reduce inflammatory cyto-kines after traumatic brain injury. Exp Neurol 2004;189:404–412.

102. Bauer ME, Jeckel CM, Luz C. The role of stress factors duringaging of the immune system. Ann N Y Acad Sci 2009;1153:139–152.

103. Zampieri S, Mellon SH, Butters TD et al. Oxidative stress inNPC1 deficient cells: protective effect of allopregnanolone.J Cell Mol Med 2008;13:3786–3796.

104. Hu Y, Cardounel A, Gursoy E, Anderson P, Kalimi M. Anti-stress effects of dehydroepiandrosterone: protection of ratsagainst repeated immobilization stress-induced weight loss,glucocorticoid receptor production, and lipid peroxidation.Biochem Pharmacol 2000;59:753–762.

105. Rupprecht R. Neuroactive steroids: mechanisms of action andneuropsychopharmacological properties. Psychoneuroendocri-nology 2003;28:139–168.

106. Wolkowitz OM, Brizendine L, Reus VI. The role ofdehydroepiandrosterone (DHEA) in psychiatry. Psychiatr Ann2000;30:123–128.

107. Raison CL, Miller AH. The neuroimmunology of stress anddepression. Semin Clin Neuropsychiatry 2001;6:277–294.

335Review: Do Stress and Depression Accelerate Cell Aging?

Depression and Anxiety

Page 10: Depression gets old fast: do stress and depression accelerate cell aging?

108. Schiepers OJ, Wichers MC, Maes M. Cytokines and majordepression. Prog Neuropsychopharmacol Biol Psychiatry2005;29:201–217.

109. O’Brien SM, Scott LV, Dinan TG. Cytokines: abnormalities inmajor depression and implications for pharmacological treat-ment. Hum Psychopharmacol 2004;19:397–403.

110. Mercanoglu G, Safran N, Uzun H, Eroglu L. Chronicemotional stress exposure increases infarct size in rats: the roleof oxidative and nitrosative damage in response to sympathetichyperactivity. Methods Find Exp Clin Pharmacol 2008;30:745–752.

111. Kiecolt-Glaser JK, Glaser R. Depression and immune function:central pathways to morbidity and mortality. J Psychosom Res2002;53:873–876.

112. Dhabhar FS, Burke HM, Epel ES et al. Low serum IL-10concentrations and loss of regulatory association between IL-6and IL-10 in adults with major depression. J Psychiatr Res2009;43:962–969.

113. Dowlati Y, Herrmann N, Swardfager W et al. A meta-analysis ofcytokines in major depression. Biol Psychiatry 2010;67:446–457.

114. Simon NM, McNamara K, Chow CW et al. A detailedexamination of cytokine abnormalities in major depressivedisorder. Eur Neuropsychopharmacol 2008;18:230–233.

115. Song C, Halbreich U, Han C, Leonard BE, Luo H. Imbalancebetween pro- and anti-inflammatory cytokines, and betweenTh1 and Th2 cytokines in depressed patients: the effect ofelectroacupuncture or fluoxetine treatment. Pharmacopsychiatry2009;42:182–188.

116. Kaur K, Sharma AK, Dhingra S, Singal PK. Interplay of TNF-alpha and IL-10 in regulating oxidative stress in isolated adultcardiac myocytes. J Mol Cell Cardiol 2006;41:1023–1030.

117. Pickering M, O’Connor JJ. Pro-inflammatory cytokines and theireffects in the dentate gyrus. Prog Brain Res 2007;163:339–354.

118. Ekdahl CT, Claasen JH, Bonde S, Kokaia Z, Lindvall O.Inflammation is detrimental for neurogenesis in adult brain.Proc Natl Acad Sci USA 2003;100:13632–13637.

119. Wichers M, Maes M. The psychoneuroimmuno-pathophysiol-ogy of cytokine-induced depression in humans. Int J Neurop-sychopharmacol 2002;5:375–388.

120. Chourbaji S, Urani A, Inta I et al. IL-6 knockout mice exhibitresistance to stress-induced development of depression-likebehaviors. Neurobiol Dis 2006;23:587–594.

121. Marsland AL, Gianaros PJ, Abramowitch SM, Manuck SB,Hariri AR. Interleukin-6 covaries inversely with hippocampalgrey matter volume in middle-aged adults. Biol Psychiatry2008;64:484–490.

122. Miller AH, Maletic V, Raison CL. Inflammation and itsdiscontents: the role of cytokines in the pathophysiology ofmajor depression. Biol Psychiatry 2009;65:732–741.

123. Davis T, Kipling D. Werner syndrome as an example ofinflamm-aging: possible therapeutic opportunities for a proger-oid syndrome? Rejuvenation Res 2006;9:402–407.

124. McIntosh LJ, Sapolsky RM. Glucocorticoids may enhanceoxygen radical-mediated neurotoxicity. Neurotoxicology1996;17:873–882.

125. Joseph JA, Shukitt-Hale B, Casadesus G, Fisher D. Oxidativestress and inflammation in brain aging: nutritional considera-tions. Neurochem Res 2005;30:927–935.

126. Houben JM, Moonen HJ, van Schooten FJ, Hageman GJ.Telomere length assessment: biomarker of chronic oxidativestress? Free Radic Biol Med 2008;44:235–246.

127. Ng F, Berk M, Dean O, Bush AI. Oxidative stress in psychiatricdisorders: evidence base and therapeutic implications. Int JNeuropsychopharmacol 2008;11:851–876.

128. Cumurcu BE, Ozyurt H, Etikan I, Demir S, Karlidag R. Totalantioxidant capacity and total oxidant status in patients withmajor depression: impact of antidepressant treatment. Psychia-try Clin Neurosci 2009;63:639–645.

129. Irie M, Asami S, Nagata S, Miyata M, Kasai H. Relationshipsbetween perceived workload, stress and oxidative DNA damage.Int Arch Occup Environ Health 2001;74:153–157.

130. Sarandol A, Sarandol E, Eker SS, Erdinc S, Vatansever E,Kirli S. Major depressive disorder is accompanied with oxidativestress: short-term antidepressant treatment does not alteroxidative-antioxidative systems. Hum Psychopharmacol 2007;22:67–73.

131. Szuster-Ciesielska A, Slotwinska M, Stachura A et al. Acceler-ated apoptosis of blood leukocytes and oxidative stress in bloodof patients with major depression. Prog NeuropsychopharmacolBiol Psychiatry 2008;32:686–694.

132. Zafir A, Ara A, Banu N. Invivo antioxidant status: a putativetarget of antidepressant action. Prog NeuropsychopharmacolBiol Psychiatry 2009;33:220–228.

133. Shen HM, Pervaiz S. TNF receptor superfamily-induced celldeath: redox-dependent execution. FASEB J 2006;20:1589–1598.

134. Tsirpanlis G, Chatzipanagiotou S, Boufidou F et al. Serumoxidized low-density lipoprotein is inversely correlated totelomerase activity in peripheral blood mononuclear cells ofhaemodialysis patients. Nephrology (Carlton) 2006;11:506–509.

135. Kapczinski F, Frey BN, Andreazza AC, Kauer-Sant’Anna M,Cunha AB, Post RM. Increased oxidative stress as a mechanismfor decreased BDNF levels in acute manic episodes. Rev BrasPsiquiatr 2008;30:243–245.

136. Grant MM, Barber VS, Griffiths HR. The presence ofascorbate induces expression of brain derived neurotrophicfactor in SH-SY5Y neuroblastoma cells after peroxide insult,which is associated with increased survival. Proteomics2005;5:534–540.

137. Groves JO. Is it time to reassess the BDNF hypothesis ofdepression? Mol Psychiatry 2007;12:1079–1088.

138. Li Y, Luikart BW, Birnbaum S et al. TrkB regulateshippocampal neurogenesis and governs sensitivity to antide-pressive treatment. Neuron 2008;59:399–412.

139. Chen B, Dowlatshahi D, MacQueen GM, Wang J-F, Young LT.Increased hippocampal BDNF immunoreactivity in subjectstreated with antidepressant medication. Biol Psychiatry2001;50:260–265.

140. Hashimoto K, Shimizu E, Iyo M. Critical role of brain-derivedneurotrophic factor in mood disorders. Brain Res Brain Res Rev2004;45:104–114.

141. Santarelli L, Saxe M, Gross C, Surget A, Battaglia F, Dulawa S.Requirement of hippocampal neurogenisis for the behavioraleffects of antidepressants. Science 2003;301:805–809.

142. Joosten EA, Houweling DA. Local acute application of BDNFin the lesioned spinal cord anti-inflammatory and anti-oxidanteffects. Neuroreport 2004;15:1163–1166.

143. Markham A, Cameron I, Franklin P, Spedding M. BDNFincreases rat brain mitochondrial respiratory coupling at complexI, but not complex II. Eur J Neurosci 2004;20:1189–1196.

144. Nitta A, Ohmiya M, Sometani A et al. Brain-derivedneurotrophic factor prevents neuronal cell death induced bycorticosterone. J Neurosci Res 1999;57:227–235.

145. Fu W, Lu C, Mattson MP. Telomerase mediates the cellsurvival-promoting actions of brain-derived neurotrophic factorand secreted amyloid precursor protein in developing hippo-campal neurons. J Neurosci 2002;22:10710–10719.

336 Wolkowitz et al.

Depression and Anxiety

Page 11: Depression gets old fast: do stress and depression accelerate cell aging?

146. Blackburn EH, Greider CW, Szostak JW. Telomeres andtelomerase: the path from maize, tetrahymena and yeast tohuman cancer and aging. Nat Med 2006;12:1133–1138.

147. von Zglinicki T. Oxidative stress shortens telomeres. TrendsBiochem Sci 2002;27:339–344.

148. Aviv A. Telomeres and human aging: facts and fibs. Sci AgingKnowledge Environ 2004;2004:pe43.

149. Simon NM, Smoller JW, McNamara KL et al. Telomereshortening and mood disorders: preliminary support for achronic stress model of accelerated aging. Biol Psychiatry 2006;60:432–435.

150. Choi J, Fauce SR, Effros RB. Reduced telomerase activity inhuman T lymphocytes exposed to cortisol. Brain Behav Immun2008;22:600–605.

151. Fuster JJ, Andres V. Telomere biology and cardiovasculardisease. Circ Res 2006;99:1167–1180.

152. Samani NJ, Boultby R, Butler R, Thompson JR, Goodall AH.Telomere shortening in atherosclerosis. Lancet 2001;358:472–473.

153. Serrano AL, Andres V. Telomeres and cardiovascular disease:does size matter? Circ Res 2004;94:575–584.

154. Valdes AM, Richards JB, Gardner JP et al. Telomerelength in leukocytes correlates with bone mineral densityand is shorter in women with osteoporosis. Osteoporos Int2007;18:1203–1210.

155. Valdes AM, Deary IJ, Gardner J et al. Leukocyte telomerelength is associated with cognitive performance in healthywomen. Neurobiol Aging (in press).

156. Epel ES, Merkin SS, Cawthon R et al. The rate of leukocytetelomere shortening predicts mortality from cardiovasculardisease in elderly men. Aging 2009;1:81–88.

157. Cawthon RM, Smith KR, O’Brien E, Sivatchenko A,Kerber RA. Association between telomere length in blood andmortality in people aged 60 years or older. Lancet 2003;361:393–395.

158. Park JI, Venteicher AS, Hong JY et al. Telomerase modulatesWnt signalling by association with target gene chromatin.Nature 2009;460:66–72.

159. Calado RT, Chen J. Telomerase: not just for the elongation oftelomeres. Bioessays 2006;28:109–112.

160. Damjanovic AK, Yang Y, Glaser R et al. Accelerated telomereerosion is associated with a declining immune function ofcaregivers of Alzheimer’s disease patients. J Immunol 2007;179:4249–4254.

161. Fitzpatrick AL, Kronmal RA, Gardner JP et al. Leukocytetelomere length and cardiovascular disease in the cardiovascularhealth study. Am J Epidemiol 2007;165:14–21.

162. Tyrka AR, Price LH, Kao HT, Porton B, Marsella SA,Carpenter LL. Childhood maltreatment and telomere short-ening: preliminary support for an effect of early stress oncellular aging. Biol Psychiatry 2010;67:531–534.

163. Cherkas LF, Aviv A, Valdes AM et al. The effects of social statuson biological aging as measured by white-blood-cell telomerelength. Aging Cell 2006;5:361–365.

164. Roux AV, Ranjit N, Jenny NS et al. Race/ethnicity and telomerelength in the multi-ethnic study of atherosclerosis. Aging Cell2009;8:251–257.

165. Hunt SC, Chen W, Gardner JP et al. Leukocyte telomeres arelonger in African Americans than in whites: the national heart,lung, and blood institute family heart study and the Bogalusaheart study. Aging Cell 2008;7:451–458.

166. Cherkas LF, Hunkin JL, Kato BS et al. The association betweenphysical activity in leisure time and leukocyte telomere length.Arch Intern Med 2008;168:154–158.

167. Atkinson SP, Keith WN. Epigenetic control of cellularsenescence in disease: opportunities for therapeutic interven-tion. Expert Rev Mol Med 2007;9:1–26.

168. Blasco MA. The epigenetic regulation of mammalian telomeres.Nat Rev Genet 2007;8:299–309.

169. Szyf M, Weaver I, Meaney M. Maternal care, the epigenomeand phenotypic differences in behavior. Reprod Toxicol 2007;24:9–19.

170. Licinio J, Wong ML. The role of inflammatory mediators inthe biology of major depression: central nervous systemcytokines modulate the biological substrate of depressive symp-toms, regulate stress-responsive systems, and contribute toneurotoxicity and neuroprotection. Mol Psychiatry 1999;4:317–327.

171. Brustolim D, Ribeiro-dos-Santos R, Kast RE, Altschuler EL,Soares MB. A new chapter opens in anti-inflammatorytreatments: the antidepressant bupropion lowers production oftumor necrosis factor-alpha and interferon-gamma in mice. IntImmunopharmacol 2006;6:903–907.

172. Barden N, Reul JM, Holsboer F. Do antidepressants stabilizemood through actions on the hypothalamic-pituitary-adrenocortical system? Trends Neurosci 1995;18:6–11.

173. Kim YK, Na KS, Shin KH, Jung HY, Choi SH, Kim JB.Cytokine imbalance in the pathophysiology of majordepressive disorder. Prog Neuropsychopharmacol Biol Psychia-try 2007;30:1044–1053.

174. Maes M, Yirmyia R, Noraberg J et al. The inflammatory andneurodegenerative (I&ND) hypothesis of depression: leads forfuture research and new drug developments in depression.Metab Brain Dis 2009;24:27–53.

175. Duman RS. Role of neurotrophic factors in the etiology andtreatment of mood disorders. Neuromolecular Med 2004;5:11–25.

176. Sen S, Duman R, Sanacora G. Serum brain-derived neuro-trophic factor, depression, and antidepressant medications:meta-analyses and implications. Biol Psychiatry 2008;64:527–532.

177. Khanzode SD, Dakhale GN, Khanzode SS, Saoji A, Palasodkar R.Oxidative damage and major depression: the potential antiox-idant action of selective serotonin re-uptake inhibitors. RedoxRep 2003;8:365–370.

178. Hashioka S, McGeer PL, Monji A, Kanba S. Anti-inflammatoryeffects of antidepressants: possibilities for preventives againstAlzheimer’s disease. Cent Nerv Syst Agents Med Chem 2009;9:12–19.

179. Eren I, Naziroglu M, Demirdas A. Protective effects oflamotrigine, aripiprazole and escitalopram on depression-induced oxidative stress in rat brain. Neurochem Res 2007;32:1188–1195.

180. Eren I, Naziroglu M, Demirdas A et al. Venlafaxine modulatesdepression-induced oxidative stress in brain and medulla of rat.Neurochem Res 2007;32:497–505.

181. Holsboer F, Ising M. Central CRH system in depression andanxiety—evidence from clinical studies with CRH1 receptorantagonists. Eur J Pharmacol 2008;583:350–357.

182. Ornish D, Lin J, Daubenmier J et al. Increased telomeraseactivity and comprehensive lifestyle changes: a pilot study.Lancet Oncol 2008;9:1048–1057.

183. Epel E, Daubenmier J, Moskowitz JT, Folkman S, Blackburn E.Can meditation slow rate of cellular aging? Cognitive stress,mindfulness, and telomeres. Ann N Y Acad Sci 2009;1172:34–53.

184. Belanoff J, Schatzberg AF. Glucocorticoid antagonists. Neuro-psychopharmacology 2000;23:S56.

337Review: Do Stress and Depression Accelerate Cell Aging?

Depression and Anxiety

Page 12: Depression gets old fast: do stress and depression accelerate cell aging?

185. Wolkowitz OM, Reus VI. Treatment of depression withantiglucocorticoid drugs. Psychosom Med 1999;61:698–711.

186. Gallagher P, Malik N, Newham J, Young AH, Ferrier IN,Mackin P. Antiglucocorticoid treatments for mood disorders.Cochrane Database Syst Rev 2008(1):CD005168.

187. McIntyre RS, Soczynska JK, Woldeyohannes HO et al.Thiazolidinediones: novel treatments for cognitive deficits inmood disorders? Expert Opin Pharmacother 2007;8:1615–1628.

188. Rasgon N, Jarvik L. Insulin resistance, affective disorders, andAlzheimer’s disease: review and hypothesis. J Gerontol A BiolSci Med Sci 2004;59:178–183. Discussion 184–192.

189. Rasgon NL, Kenna HA. Insulin resistance in depressivedisorders and Alzheimer’s disease: revisiting the missing linkhypothesis. Neurobiol Aging 2005;26:103–107.

190. Mathew SJ, Keegan K, Smith L. Glutamate modulators as novelinterventions for mood disorders. Rev Bras Psiquiatr 2005;27:243–248.

191. Maeng S, Zarate Jr CA. The role of glutamate in mooddisorders: results from the ketamine in major depression studyand the presumed cellular mechanism underlying its anti-depressant effects. Curr Psychiatry Rep 2007;9:467–474.

192. Skolnick P, Popik P, Trullas R. Glutamate-based antidepres-sants: 20 years on. Trends Pharmacol Sci 2009;30:563–569.

193. Zarate CA, Manji HK. Riluzole in psychiatry: a systematicreview of the literature. Expert Opin Drug Metab Toxicol2008;4:1223–1234.

194. Sanacora G, Zarate CA, Krystal JH, Manji HK. Targeting theglutamatergic system to develop novel, improved therapeuticsfor mood disorders. Nat Rev Drug Discov 2008;7:426–437.

195. Paul IA. Antidepressant activity and calcium signaling cascades.Hum Psychopharmacol 2001;16:71–80.

196. Dubovsky SL, Buzan R, Thomas M, Kassner C, Cullum CM.Nicardipine improves the antidepressant action of ECT butdoes not improve cognition. J Ect 2001;17:3–10.

197. Cocchi P, Silenzi M, Calabri G, Salvi G. Antidepressant effectof vitamin C. Pediatrics 1980;65:862–863.

198. Dunn AL, Trivedi MH, Kampert JB, Clark CG, ChamblissHO. Exercise treatment for depression: efficacy and doseresponse. Am J Prev Med 2005;28:1–8.

199. Blake H, Mo P, Malik S, Thomas S. How effective are physicalactivity interventions for alleviating depressive symptoms inolder people? A systematic review. Clin Rehabil 2009;23:873–887.

200. Mead GE, Morley W, Campbell P, Greig CA, McMurdo M,Lawlor DA. Exercise for depression. Cochrane Database SystRev 2009(3):CD004366.

201. Strohle A, Stoy M, Graetz B et al. Acute exercise amelioratesreduced brain-derived neurotrophic factor in patients withpanic disorder. Psychoneuroendocrinology 2010;35:364–368.

202. Kishino A, Katayama N, Ishige Y et al. Analysis of effectsand pharmacokinetics of subcutaneously administered BDNF.Neuroreport 2001;12:1067–1072.

203. Zhang Y, Pardridge WM. Conjugation of brain-derivedneurotrophic factor to a blood-brain barrier drug targetingsystem enables neuroprotection in regional brain ischemiafollowing intravenous injection of the neurotrophin. Brain Res2001;889:49–56.

204. Egleton RD, Davis TP. Development of neuropeptide drugsthat cross the blood-brain barrier. NeuroRx 2005;2:44–53.

205. Effros RB. Telomerase induction in Tcells: a cure for aging anddisease? Exp Gerontol 2007;42:416–420.

206. Fossel M. Telomerase and the aging cell: implications forhuman health. J Am Med Assoc 1998;279:1732–1735.

207. Soczynska JK, Kennedy SH, Goldstein BI, Lachowski A,Woldeyohannes HO, McIntyre RS. The effect of tumornecrosis factor antagonists on mood and mental health-associated quality of life: novel hypothesis-driven treatmentsfor bipolar depression? Neurotoxicology 2009;30:497–521.

208. Mendlewicz J, Kriwin P, Oswald P, Souery D, Alboni S,Brunello N. Shortened onset of action of antidepressants inmajor depression using acetylsalicylic acid augmentation: apilot open-label study. Int Clin Psychopharmacol 2006;21:227–231.

209. Nishida A, Miyaoka T, Inagaki T, Horiguchi J. New approachesto antidepressant drug design: cytokine-regulated pathways.Curr Pharm Des 2009;15:1683–1687.

210. Tyring S, Gottlieb A, Papp K et al. Etanercept and clinicaloutcomes, fatigue, and depression in psoriasis: double-blindplacebo-controlled randomised phase III trial. Lancet 2006;367:29–35.

211. Berthold-Losleben M, Heitmann S, Himmerich H. Anti-inflammatory drugs in psychiatry. Inflamm Allergy DrugTargets 2009;8:266–276.

212. Krystal JH, Sanacora G, Blumberg H et al. Glutamate andGABA systems as targets for novel antidepressnat and mood-stabilizing treatments. Mol Psychiatry 2002;7:S71–S80.

213. Heim C, Newport DJ, Mletzko T, Miller AH, Nemeroff CB.The link between childhood trauma and depression: insightsfrom HPA axis studies in humans. Psychoneuroendocrinology2008;33:693–710.

214. Tyrka AR, Price LH, Kao HT, Porton B, Marsella SA,Carpenter LL. Telomere shortening and reports of childhoodmaltreatment in healthy adults: a pilot study. The AnnualConference of the American College of Neuropsychopharma-cology. Boca Raton, FL; 2008.

215. Anda RF, Felitti VJ, Bremner JD et al. The enduring effects ofabuse and related adverse experiences in childhood : a conver-gence of evidence from neurobiology and epidemiology. EurArch Psychiatry Clin Neurosci 2006;256:174–186.

338 Wolkowitz et al.

Depression and Anxiety