Você está na página 1de 20

Neuroscience 135 (2005) 659 – 678

REVIEW
THE PATHOGENESIS OF CLINICAL DEPRESSION: STRESSOR- AND
CYTOKINE-INDUCED ALTERATIONS OF NEUROPLASTICITY
S. HAYLEY,a* M. O. POULTER,a Z. MERALIb AND Contents
H. ANISMANa,b Neurochemical underpinning of depressive illness 660
a
Institute of Neuroscience, Carleton University, 1125 Colonel By Drive,
Neurotrophic factors in depression 662
Ottawa, Ontario, Canada K1S 5B6 Neuroplasticity and depression 663
b
Growth factors in relation to depression 663
University of Ottawa Institute of Mental Health Research, Department of Neurogenesis and hippocampal atrophy in depression 664
Psychology, Department of Psychiatry, Ottawa, Ontario, Canada K1Z 7K4 Changes of neuroplasticity in depression 665
Neurotrophins and cytokines 666
Abstract—Stressful events promote neurochemical changes Stress– cytokine– depression connection 667
that may be involved in the provocation of depressive disor- Cytokine effects on neurochemical activity 667
der. In addition to neuroendocrine substrates (e.g. cortico- Proactive effects of cytokines and stressors 668
tropin releasing hormone, and corticoids) and central neuro- Cytokines in depression 668
transmitters (serotonin and GABA), alterations of neuronal Animal models: sickness and depression 668
plasticity or even neuronal survival may play a role in depres- Clinical evidence for cytokines in depression 669
sion. Indeed, depression and chronic stressor exposure typ- Neurodegenerative aspects of depression: cytokine
ically reduce levels of growth factors, including brain-derived involvement 670
neurotrophic factor and anti-apoptotic factors (e.g. bcl-2), as Concluding remarks 670
well as impair processes of neuronal branching and neuro- Acknowledgments 672
genesis. Although such effects may result from elevated cor- References 672
ticoids, they may also stem from activation of the inflamma-
tory immune system, particularly the immune signaling cyto-
kines. In fact, several proinflammatory cytokines, such as Appreciation of the processes underlying depressive dis-
interleukin-1, tumor necrosis factor-␣ and interferon-␥, influ- orders has increased considerably over the past decades,
ence neuronal functioning through processes involving apo- both in terms of the neurochemical substrates for the dis-
ptosis, excitotoxicity, oxidative stress and metabolic de- order as well as some of the psychosocial factors that
rangement. Support for the involvement of cytokines in de-
promote pathology. Few investigators would argue against
pression comes from studies showing their elevation in
severe depressive illness and following stressor exposure,
the view that stressful events play a prominent role in the
and that cytokine immunotherapy (e.g. interferon-␣) elicited provocation of depression, particularly in the presence of
depressive symptoms that were amenable to antidepressant genetic vulnerability factors (Caspi et al., 2003). Likewise,
treatment. It is suggested that stressors and cytokines share a constellation of psychosocial factors (e.g. coping ability,
a common ability to impair neuronal plasticity and at the previous stressful experiences, familial factors) moderates
same time altering neurotransmission, ultimately contribut- the effects of stressful events on depression (Anisman and
ing to depression. Thus, depressive illness may be consid- Matheson, 2005). It is assumed that these factors come
ered a disorder of neuroplasticity as well as one of neuro-
together to affect neuronal processes, culminating in the
chemical imbalances, and cytokines may act as mediators of
both aspects of this illness. © 2005 IBRO. Published by provocation or exacerbation of behavioral pathology.
Elsevier Ltd. All rights reserved. Uncovering the specific neurochemical mechanisms
subserving depressive illnesses is exceedingly complex,
Key words: BDNF, CRH, interleukin, 5-HT, neurogenesis, in- particularly as depression is often considered to be a bio-
flammatory. chemically heterogeneous disorder. Indeed, subtypes of
*Corresponding author. Tel: ⫹1-613-520-2600x6314; fax: ⫹1-613- depression exist (e.g. typical vs atypical depression; recur-
520-4052. rent depression; dysthymic disorder) that may involve sev-
E-mail address: shayley@ccs.carleton.ca (S. Hayley).
Abbreviations: ACTH, adrenocorticotropin; AVP, arginine vasopressin;
eral common neurochemical features, but also several
BDNF, brain-derived neurotrophic factor; BDZ, benzodiazepine; CREB, distinct differences (Gold and Chrousos, 2002; Griffiths et
cAMP responsive element binding protein; CRH, corticotropin releasing al., 2000). Furthermore, just as individuals diagnosed with
hormone; CSF, cerebral spinal fluid; ERK, extracellular signal-regulated a particular form of depression may present with some-
kinase; HPA, hypothalamic–pituitary–adrenal; IDO, indoleamine 2,3-di-
oxygenase; IFN, interferon; IL-1, interleukin-1; IL-1ra, interleukin-1 recep- what different symptoms, there is reason to believe that the
tor antagonist; JNK, c-Jun N-terminal kinase; LPS, lipopolysaccharide; processes subserving depression in one individual may
MAP, mitogen-activated protein; NE, norepinephrine; NT-3, neurotro- differ markedly from those underlying depression in a sec-
phin-3; PFC, prefrontal cortex; PKA, cAMP binding to protein kinase A;
PTSD, post traumatic stress disorder; SSRI, selective serotonin reuptake ond individual. The view that multiple neurochemical alter-
inhibitor; TNF-␣, tumor necrosis factor-␣; 5-HT, serotonin. ations may instigate depression may account for some of
0306-4522/05$30.00⫹0.00 © 2005 IBRO. Published by Elsevier Ltd. All rights reserved.
doi:10.1016/j.neuroscience.2005.03.051

659
660 S. Hayley et al. / Neuroscience 135 (2005) 659 – 678

the interindividual variability of symptoms presented as and tryptophan hydroxylase gene polymorphisms (Faludi
well as the variability, and frequent unpredictability in the et al., 2000) and increased frequency of the 5-HT trans-
efficacy of drug treatments for depression. porter gene long (L) allele (Du et al., 1999). More recent
The present review is meant to provide a framework by data indicated that a 5-HT1A repressor gene promoter in
which chronic stressful events (or acute events that are raphe cells was reduced by a promoter polymorphism.
associated with chronic rumination) may come to affect This suggested that the allele that de-represses 5-HT1A
neurochemical processes that impact on depressive ill- autoreceptor expression [G(⫺1019)] is potentially respon-
ness. Essentially, it is argued that chronic stressful events sible for the reduced 5-HT neurotransmission, hence pre-
may instigate the activation of a cascade of neurochemical disposing individuals to depression and suicide (Lemonde
and/or inflammatory factors that ultimately affect neuronal et al., 2003).
functioning and/or survival, thus influencing depression. In The available data have not uniformly supported a role
particular, in this working model chronic stressors or cen- for 5-HT dysfunctions in depression (Anguelova et al.,
tral cytokine activation may influence limbic corticotropin 2003a,b; Stockmeier, 2003), as several studies failed to
releasing hormone (CRH) functioning and the subsequent show a relationship between suicide/depression and 5-HT
provocation of monoamine disturbances (particularly re- functioning (Lowther et al., 1997; Meyer et al., 1999).
duced serotonin (5-HT) turnover and altered GABA func- Further, analyses controlling for age of subject, postmor-
tioning) that favor the emergence of depressive states. The tem interval (from the time of suicide), and the time of
CRH, 5-HT and cytokine changes are linked to alterations tissue storage, revealed that differences of 5-HT1A recep-
of growth/anti-apoptotic factors (e.g. brain-derived neuro- tors between depressed suicides and controls were not
trophic factor (BDNF) and bcl-2), which may, in turn, affect evident (Stockmeier, 2003; Stockmeier et al., 1997). Over-
neuroplasticity, ultimately resulting in depression. It is also all, it seems that the data from postmortem analyses at-
suggested that parallel pathways exist that could influence tempting to specify the specific neurochemical distur-
depressive outcomes. Although the individual pathways bances related to depression have been inconsistent
linking depression to CRH, cytokines, and growth factors is (Gross-Isseroff et al., 1998). Certainly, multiple neuro-
well-trod ground, in this review we integrate these pro- transmitters have been implicated in depression, and it is
cesses with neurotransmitter systems (5-HT, GABA) that possible that some of the inconsistencies reported reflect
have been implicated in depressive illness. the complex interactions that may occur between transmit-
ter systems.
Neurochemical underpinning of depressive illness As depicted in Fig. 1, there are considerable data
Depressive illness has long been associated with distur- focusing on the possibility that stressor-elicited hypotha-
bances of brain 5-HT, norepinephrine (NE) and dopamine lamic–pituitary–adrenal (HPA) hormones (CRH, adreno-
activity, variations of 5-HT1A and 5-HT2 receptors, as well corticotropin (ACTH), cortisol) are associated with depres-
as ␣1-NE or ␤-NE receptors (Maes and Meltzer, 1995; sion, as might CRH within extrahypothalamic sites, notably
Schatzberg and Schildkraut, 1995). The data concerning the amygdala, PFC and locus coeruleus (Holsboer, 2003;
5-HT variations in depression have probably been the Nemeroff, 1996). There is reason to believe that CRH and
most widely studied. Evidence in favor of the perspective cortisol are not simply bystanders, but play a provocative
that 5-HT is involved in depression comes not only from role in depression. Consistent with this position, circulating
studies showing that pharmacological manipulation of cortisol levels, as well as CRH concentrations in cerebro-
5-HT attenuates depression (Blier, 2003), but also from spinal fluid are elevated among depressed patients (Arato
postmortem and cerebral spinal fluid (CSF) studies indi- et al., 1989; Arborelius et al., 1999; Banki et al., 1992;
cating low levels of 5-HT and its metabolite, 5-hydroxyin- Heuser et al., 1998; Nemeroff et al., 1984; Wong et al.,
doleacetic acid (5-HIAA), in suicide victims and suicide 2000). While there have also been reports indicating a
attempters. Analyses of depressed suicide brains revealed failure to detect elevated CRH within CSF (Kling et al.,
morphological and functional disturbances within specific 1991; Pitts et al., 1995), it seems that CRH CSF levels
aspects of the prefrontal cortex (PFC; e.g. dorsomedial were particularly elevated in those patients who were
and orbital cortex), as part of a wider system of neuronal dexamethasone nonsuppressors (Roy et al., 1987). Fur-
processes that may contribute to the emergence of major ther, pharmacotherapy or electroconvulsive therapy nor-
depressive illness (Anguelova et al., 2003a,b; Drevets, malized the high levels of CSF CRH associated with de-
2001). The disturbances that characterize the suicide brain pression (Heuser et al., 1998; Nemeroff et al., 1991).
have included reductions of neurons and glial cells (Ra- The data concerning concentration of CRH and its
jkowska, 2000), a reduction of cyclic AMP (cAMP) binding receptors in the brain of depressed patients are relatively
to protein kinase A (PKA), and a decrease of PKA activity limited. Nevertheless, CRH mRNA expression and/or CRH
(Dwivedi et al., 2002). Depression was also associated immunoreactivity was reported to be elevated in hypotha-
with an increase of 5HT1A and 5HT2A receptors (Arango et lamic nuclei, locus coeruleus and raphe nuclei of de-
al., 1995; Escriba et al., 2004; Mann et al., 2000; Pandey pressed suicides (Austin et al., 2003; Bissette et al., 2003;
et al., 2002; Zanardi et al., 2001), as well as a decrease of Raadsheer et al., 1994, 1995). Despite the frequent as-
5-HT transporter sites (Arango et al., 1995; Faludi et al., sumption that disturbances of frontal cortical processes
2000; Mann et al., 2000). It also appeared that depressed- may be involved in depressive illnesses, data concerning
suicide brains were associated with altered 5HT2A receptor CRH-related changes in these regions have been sparse
S. Hayley et al. / Neuroscience 135 (2005) 659 – 678 661

Fig. 1. Working model depicting potential routes by which chronic stressors could impact depressive state. A chronic stressor could potentially
influence cytokine functioning (note that acutely administered stressors increase cytokine activity, whereas chronic stressors, as depicted in the figure,
functionally reduce cytokine activity), excite CRH release, and inhibit BDNF (black lines); CRH may reciprocally modulate GABAA receptor and inhibit
5-HT neuronal functioning within the PFC, hippocampus and amygdala (depicted by green lines), hence leading to depression; 5-HT may influence
levels of growth and anti-apoptotic factors (BDNF and bcl-2, respectively) favoring impaired cellular plasticity and survival, hence promoting depression
(purple lines); cytokine release, which activate the HPA axis, thereby promoting hormonal variations aligned with depression (red lines); (d: depicted
by orange lines) cytokine release may directly impact cell survival or negatively impinge upon neuroplasticity through apoptotic, oxidative or excitotoxic
processes (orange lines); cytokine activity leading to provocation of toxic metabolites, such as IDO which may directly impair 5-HT functioning, or result
in increased accumulation of kynurenine metabolites (3-OH-kynurenine and hippocampal NMDA), which would then act in a neurotoxic capacity (blue
lines), hence promoting behavioral disturbances. These multiple pathways are not considered to be exclusive of one another.
and inconsistent. While Nemeroff et al. (1988) reported in this regard (Dautzenberg and Hauger, 2002; Nemeroff,
reduced CRH binding sites within the frontal cortex of 1996; Reul and Holsboer, 2002). Data such as these have
depressed suicides, neither Hucks et al. (1997) nor Leake been instrumental in prompting the development of thera-
et al. (1990) observed such effects. However, it has been peutic agents for depression that targeted CRH functioning
found that alterations of CRH levels and receptor mRNA (Nemeroff, 1996; Holsboer, 2003).
expression may occur in selective aspects of the PFC Despite the impressive evidence suggesting a relation-
(Merali et al., 2004a). Specifically, among depressed sui- ship between suicide/depression and both 5-HT and CRH
cide victims CRH levels were elevated in the right fronto- functioning, we emphasize again that the available data
polar (FPC) and dorsomedial PFC (only tissue from the have not been unambiguous. Besides the negative find-
right side was assessed in that work), whereas mRNA for
ings reported by Hucks et al. (1997) and Leake et al.
CRH1, but not CRH2 receptors, was reduced. Presumably,
(1990), it was recently reported (Sibille et al., 2004) that
the CRH1 desensitization develops with excessive CRH
suicide was not associated with unique gene expression
activation (Aguilera et al., 2001; Pozzoli et al., 1996). In-
levels within two regions of the PFC (dorsomedial and
deed, it was reported that acute stressors increased CRH
receptor mRNA expression and CRH content in both post- orbital) with respect to CRH, CRH1, CRH2, CRH binding
mortem and in vivo analyses of rodent central amygdala protein, as well as several 5-HT receptors, tryptophan
(Cook, 2001; Merali et al., 1998, 2004b; Merlo et al., 1995). hydroxylase, and the 5-HT transporter. The investigators
Moreover, in chronically stressed animals, a condition that could not readily determine the source for the inconsisten-
may be more similar to the sustained distress of severe cies between their findings and those of numerous other
depression, a decrease of CRH1 expression was provoked studies, but raised the possibility that either these aspects
within PFC (Iredale et al., 1996). Together, these data of the PFC were not related to depression/suicide, or the
support a role for CRH elevations in depression/suicide, genomic differences from controls were below the detec-
and suggest that the CRH1 subtype is particularly pertinent tion limit for the methodology used.
662 S. Hayley et al. / Neuroscience 135 (2005) 659 – 678

In addition to CRH involvement in depression, there is 2001), coupled with its influence on nucleus accumbens
reason to believe that GABA may also contribute in this functioning (a region that may be fundamental in the an-
respect, particularly as CRH and 5-HT may influence hedonic features of depression; Stevenson et al., 2003),
GABA activity (Sibille et al., 2000; Steiger and Russek, we assessed the expression of the ␣ (␣1, ␣2, ␣3, ␣4, ␣5), ␦
2004; Tan et al., 2004), thereby influencing mood disor- and ␥2 receptor subunits that compose the GABAA recep-
ders (e.g. Merali et al., 2004b). Indeed, several sources of tor complex in several aspects of the frontal cortex, namely
evidence have implicated GABA functioning in depressive the frontopolar, dorsomedial and ventrolateral prefrontal
illness, including (a) increased plasma and CSF GABA cortices (Merali et al., 2004a). In addition to the CRH
levels in depression, (b) the effects of GABA acting agents receptor variations described earlier, mRNA expression of
on symptoms of depression/anxiety, and (c) the influence the ␣1, ␣3, ␣4, and ␦ receptor subunits was reduced in the
of antidepressants on GABAergic interneuronal function- frontopolar region of suicide victims. Importantly, our par-
ing (Brambilla et al., 2003; Krystal et al., 2002; Petty, 1995; tial analysis of the GABAA receptor functional genome
Shiah and Yatham, 1998; Tunnicliff and Malatynska, revealed marked cross-correlations with respect to subunit
2003). The potential involvement of GABA in depression expression in cortical regions of nondepressed individuals,
was further reinforced by the finding that reciprocal inner- suggesting a high degree of coordinated gene regulation
vation exists between GABA and 5-HT within brain regions (Merali et al., 2004b). It was of particular significance that
often associated with depression, including the PFC and the GABA subunit intercorrelations were markedly attenu-
hippocampus (Brambilla et al., 2003). Yet, postmortem ated in suicide brains (with the exception of the interrela-
brain analyses concerning the involvement of this trans- tions between the ␣2, ␣3 and ␣4 subunits). As an illustrative
mitter in depression have not yielded compelling data. example, Fig. 2 shows the scatter plots between ␣1 and
While GABA levels within the PFC were inversely related each of the other subunits in frontopolar cortex of control
to severity of depression (Honig et al., 1988), conflicting suicide victims. These findings raise the possibility that the
results were reported showing that neither GABA levels GABAA receptor subunit changes, or the disturbed coordi-
(Korpi et al., 1988), GABA-related enzymes (Cheetham et nation between the GABAA receptor subunits, contribute to
al., 1988; Sherif et al., 1991), the GABA transporter (Sund- depression/suicide or are secondary to the illness/distress
man-Eriksson and Allard, 2002) nor GABAB binding in the associated with it. Other investigators have similarly ob-
frontal cortex differed between drug-free depressed sui- served coordinated GABAA receptor subunit expression
cide victims and controls (Cross et al., 1988; Sundman et (␣1, ␣2 ␣3, ␤2/␤3, ␥2, ␦) within dentate granule neurons
al., 1997). Importantly, however, GABAA/BDZ (benzodiaz- (Brooks-Kayal et al., 1999; Kern and Sieghart, 1994;
epine) binding sites were elevated in depressed suicides Mertens et al., 1993), and as the genes for these subunits
(Cheetham et al., 1988), indicating that GABAA receptor appear on different chromosomes, the subunit coordina-
subunit expression is likely changed. tion could not be attributed to factors related to shared
An interesting link between 5-HT and GABA receptors chromosomal localization (e.g. linkage effects).
has been reported, essentially indicating that 5HT1A recep- It is premature to offer strong positions concerning the
tors regulate GABAA receptor expression (Sibille et al., processes subserving the interrelations between CRH and
2000). Specifically, 5-HT1A knockout mice respond poorly 5HT receptors and the coordinated GABAA subunit ex-
to BDZ anxiolysis, an effect attributed to a loss of GABA pression. The occurrence of changes within these trans-
binding sites, and specifically to ␣1 and ␣2 GABAA subunit mitter systems may be entirely independent of one an-
decreases in the CA1 and dentate of the hippocampus. other, with one affecting a particular attribute of the wide
This link between 5-HT receptor status and GABAA recep- range of behavioral symptoms that occur in depressed
tor expression corroborates the well-known findings that individuals, while another may be associated with other
depressed patients often respond poorly to BDZ therapy characteristics of depressed suicides. It is equally possi-
(Roy-Byrne et al., 1996; Schlegel et al., 1994). ble, however, that CRH and GABA systems are related to
Inasmuch as CRH receptors regulate the responsive- one another, and together influence the profile of the de-
ness of 5-HT receptors, it is important to consider the pressed suicide. In this respect, as already indicated,
involvement of CRH in the GABA-5HT receptor relation- GABAergic acting agents affect limbic CRH mRNA expres-
ship. It seems that CRH potentiates the enhancement of sion (Gilmor et al., 2003; Stout et al., 2001), and may
GABAA receptor inhibition caused by 5-HT application, contribute to the stressor-provoked CRH response (Culli-
suggesting the occurrence of functional cross-talk between nan and Wolfe, 2000). Thus, just as CRH may influence
these receptor systems. In this regard, CRH and GABAA monoamine (both NE and 5-HT) neuronal activity, which
function/expression may be interrelated in that CRH is could potentially impact depressive states (Ruggiero et al.,
uniquely expressed in GAD positive interneurons in rat 1999), it is possible that CRH-GABA interrelations have
cortex and chronic stressors can affect GABAA receptor such an effect. Alternatively, CRH variations may instigate
expression (Cullinan and Wolfe, 2000). Likewise, GABAer- 5-HT receptor changes, which then influence frontal corti-
gic agents may affect CRH mRNA expression within limbic cal GABAA functioning (Tan et al., 2004; Yan et al., 2001).
sites (Gilmor et al., 2003; Stout et al., 2001).
Neurotrophic factors in depression
Given the important role for the PFC in subserving
cognitive processes, including hedonia and behavioral Analysis of the underpinnings of depression has increas-
planning (Kalivas and Nakamura, 1999; Tanji and Hoshi, ingly focused on deficiencies of neuroplasticity and im-
S. Hayley et al. / Neuroscience 135 (2005) 659 – 678 663

Fig. 2. Scatter plots showing relations between the ␣1 GABAA receptor subunit mRNA abundance and those of several other subunits (␣2, ␣3, ␣4, ␣5,
␦ and ␥) in the frontopolar cortex of suicide victims (right hand panels) and controls (left hand panels). Note the tight correlations evident between the
subunit abundance in control brains (most of the cross-correlations between these subunits were significant). In the suicide brains these relations were
limited (data from Merali et al., 2004, which reports all the cross-correlations between these subunits).

paired production of trophic factors (Manji et al., 2001). Neuroplasticity and depression
The target of experimental agents has included treatments
The belief that depression is strictly a disorder of neuro-
that promote cell survival such as the growth factor, BDNF,
and the anti-apoptotic protein, bcl-2 (Duman, 2004; Hashi- chemical imbalances has undergone reconsideration with
moto et al., 2004; Manji and Duman, 2001). These have the realization that some degree of impaired neuronal
received particular attention as stressors and depression survival or structural abnormalities may be evident in this
have been implicated in alterations of neuronal plasticity or disorder (Harrison, 2002; Manji et al., 2001). Essentially,
even injury, whereas antidepressants may act on path- the “depressed brain” may be unable to produce appropri-
ways associated with enhanced cellular plasticity and sur- ate adaptive neuronal responses (e.g. such as changes of
vival (see Fig. 1). In this regard, it was shown that infusion synaptic connections or dendritic branching required to
of BDNF into the dorsal raphe nucleus provoked an anti- deal with stressors or other environmental challenges),
depressant-like effect in rodents tested in the learned help- hence rendering individuals vulnerable to emotional and
lessness model of depression (Altar, 1999). The fact that cognitive disturbances. When faced with extreme, unre-
BDNF also promoted sprouting of central 5-HT neurons lenting stressors, degeneration of neurons may even occur
(Altar, 1999) supported the view that its antidepressant (Uno et al., 1994), an effect that may be exacerbated in
effect may stem from structural remodeling of these neu- depressed individuals.
rons. Likewise, infusion of BDNF or the related neurotro-
phin-3 (NT-3) protein into the hippocampus produced an- Growth factors in relation to depression. We have
tidepressant-like effects in the learned helplessness and argued that inflammatory immune system factors might
forced swim models of depression (Shirayama et al., contribute to many of the neurochemical and neuroplastic
2002). Limited information is available concerning BDNF in deficiencies associated with clinical depression. Of course,
the brain of depressed individuals. However, it was re- this is still a controversial issue and it is not even univer-
ported that mRNA and protein levels of BDNF and its sally accepted that altered neuroplasticity is a prominent
receptor (tyrosine kinase B, TrkB) were lower within the aspect of depression. In this regard, it is unclear whether
PFC and hippocampus of suicides than in age- and sex- variations of neuroplasticity play a causal role in depres-
matched controls (Dwivedi et al., 2003a). Further, varia- sive pathology or are simply a product of the chronic
tions of neurotrophins other than BDNF in human patients distress associated with ongoing depression (Duman,
were observed in that CSF levels of NT-3 were elevated in 2004; Hayley and Anisman, 2004; MacQueen et al., 2003).
depressed individuals (Hock et al., 2000). Nevertheless, several studies revealed that stressors im-
664 S. Hayley et al. / Neuroscience 135 (2005) 659 – 678

pair neuronal branching, growth and survival (Duman, (Coyle and Duman, 2003). Consistent with the view that
2004; Gould and McEwen, 1993; Manji et al., 2003), and neurogenesis may be relevant to depression, this impair-
given that stressors precipitate depression, the influence of ment was reversed by chronic antidepressant treatment or
these processes warrants continued attention. It ought to by a single session of electroconvulsive shock (Jacobs,
be underscored, as well, that the changes of neuroplastic- 2002). Human postmortem analyses likewise indicated
ity may stem from BDNF and bcl-2 variations brought that neurogenesis was diminished within the hippocampus
about by 5-HT changes that may have been induced by of patients suffering from major depression (Kempermann
stressors (see Fig. 1). Indeed, 5-HT and BDNF influence and Kronenberg, 2003). As alluded to earlier, such an
one another, such that 5-HT enhances BDNF expression, outcome could come about through several mechanisms,
and BDNF ensures 5-HT neuron survival. Thus, stressor- including the possibility that the altered turnover of 5-HT
induced variations of 5-HT may ultimately come to influ- ordinarily associated with stressors may provoke distur-
ence neuroplasticity (Mattson et al., 2004). bances of growth factors important in neurogenesis (see
As mentioned earlier, depression is a biochemically Fig. 1) (Mattson et al., 2004).
heterogeneous disorder, with multiple subtypes (e.g. dys- Reduced hippocampal volume associated with clinical
thymic and major depressive illness; typical and atypical depression may be related to the direct actions of glu-
subtypes), and alterations of plasticity may only be evident cocorticoids, which are frequently elevated in relatively
in certain forms of the disorder. These differences notwith- severe cases of depression or in response to chronic
standing, several imaging studies have revealed that hip- stressors (Sapolsky et al., 1985). Specifically, corticoids
pocampal volume was reduced among depressed pa- impair glucose transport into hippocampal pyramidal neu-
tients, and postmortem analyses indicated that this out- rons, thus resulting in an energetic compromise of these
come was directly related to illness duration (Bremner et cells (McEwen, 2001). Glucocorticoids may potentiate el-
al., 2000; Campbell et al., 2004; Sheline et al., 2003) and evations of free cytosolic calcium, and hence may either
was most evident in depressed individuals carrying the directly impair neuronal survival or render cells vulnerable
long variant of the 5-HT transporter (Frodl et al., 2004). to other insults. In addition, corticoids can exacerbate
Furthermore, symptom remission was associated with hip- these vulnerabilities through excitotoxic processes involv-
pocampal volume (and lateralization), with non-remitted ing elevations of NMDA and/or AMPA glutamate receptors
individuals showing a greater reduction than patients in (McEwen, 2001). Consistent with this perspective, adre-
remission (Frodl et al., 2004), particularly within the left nalectomy attenuated the stressor-induced increase of glu-
hippocampus (Mervaala et al., 2000; MacMaster and Ku- tamate typically observed in the hippocampus and PFC
sumakar, 2004; Vakili et al., 2000). Others have failed to (Moghaddam et al., 1994). In addition to excitotoxic pro-
find such morphological changes, but did report memory cesses, excessive levels of the hormone may contribute to
deficits (Vythilingam et al., 2004). It is also possible that free radical accumulation, largely by reducing the capacity
reduced hippocampal volume may be a risk for depres- of antioxidant enzymes (Sapolsky, 2000). Ultimately, such
sion, rather than a result of the disorder, as has been corticoid-mediated effects may lead to either hippocampal
shown in the case of post traumatic stress disorder (PTSD) neuronal demise or the regression of pyramidal neuron
(Gilbertson et al., 2002). dendritic branching (Sapolsky, 2000). Either situation
Although the mechanisms responsible for reduced hip- would elicit a reduction of hippocampal volume like that
pocampal volume have yet to be determined, a variety of observed in chronically depressed patients.
potential processes have been advanced, including re- Further support for the damaging effects of stressors
duced neurogenesis, diminished dendritic branching, as and glucocorticoids comes from studies in monkeys dem-
well as induction of excitotoxic and apoptotic death mech- onstrating that hippocampal implantation of cortisol pellets
anisms. Such effects have been linked to the excessive for 1 year provoked morphological changes in the hip-
glucocorticoid activity that is frequently evident among de- pocampal CA3/CA2 subfields, including irregular cell lay-
pressed individuals and it is possible that cytokine-medi- ers, soma shrinkage and dendritic atrophy (Sapolsky et al.,
ated neuroinflammatory processes may also be involved 1990). Likewise, in rats, intermittent restraint exacerbated
(Ma et al., 2002; Sapolsky, 2000; Stepanichev et al., the hippocampal neuronal loss, as well as expression of
2003). It is important to underscore, however, that most the degenerative marker, tau protein, seen after kainic acid
studies of altered brain architecture involved modifications treatment (Stein-Behrens et al., 1994). Moreover, chronic
of hippocampal volume. Yet, this region is only one of corticosterone treatment increased hippocampal neuronal
many that may be involved in depressive illness. To be loss, whereas glial cells displayed abnormal morphology
sure, various aspects of the PFC, amygdala and hypothal- (Sapolsky et al., 1985). The hippocampal cell loss was also
amus have been implicated in depression, but data regard- evident in response to chronic psychosocial stressors.
ing atrophy of these regions have not received equivalent Specifically, subordinate male tree shrews chronically ex-
attention (Harrison, 2002). posed to a dominant animal displayed reduced apical den-
drite length of hippocampal pyramidal neurons and fewer
Neurogenesis and hippocampal atrophy in depression. dendritic branching points (Magarinos et al., 1996). Simi-
Consistent with a stress model of depression, rodents larly, pyramidal neurons within the PFC displayed reduced
exposed to a chronic stressor regimen or corticosterone apical dendritic branching and length in response to a
treatments displayed impaired hippocampal neurogenesis chronic restraint stressor regimen (Cook and Wellman,
S. Hayley et al. / Neuroscience 135 (2005) 659 – 678 665

2004). Paralleling these findings, humans suffering pro- provoked corticosterone release (Smith et al., 1995). Inter-
tracted trauma or PTSD displayed reduced volume of the estingly, however, the reduced BDNF associated with re-
cerebral cortex as revealed in imaging studies (Grossman straint was prevented by antidepressant treatment (Duman
et al., 2002; Yehuda, 2002). Of course, as already indi- et al., 1999). An upregulation of BDNF expression was
cated, the latter data are correlational and the possibility also associated with improved performance (i.e. reduced
cannot be excluded that among individuals with a dimin- immobility) among rats in the forced swim test, a paradigm
ished hippocampus, traumatic events may be more likely that has been used to screen antidepressant agents
to result in PTSD (Gilbertson et al., 2002). (Shirayama et al., 2002). While there is certainly evidence
Although corticoids are capable of inhibiting neurogen- that stressors generally reduce hippocampal or hypotha-
esis (Manji et al., 2003), factors other than these hormones lamic BDNF levels, the observed effects may vary in a
may be responsible for the observed effects in depression. biphasic fashion with the duration of the stressor (Givalois
In particular, chronic electroshock treatment was associ- et al., 2004; Marmigere et al., 2003; Rage et al., 2002), in
ated with increased neurogenesis, even in the presence of that brief stressors of moderate severity may increase
elevated corticoid levels (Hellsten et al., 2002). It was BDNF expression, whereas more extended insults may
similarly reported that the antidepressant, fluoxetine, nor- have the opposite effect.
malized hippocampal neurogenesis in animals exposed to The mitogen activated protein (MAP) kinase signaling
inescapable electric footshock, independent of any actions pathways may be important for BDNF and bcl-2 function-
upon corticosterone (Malberg and Duman, 2003; Malberg ing (Ying et al., 2002; Eom et al., 2004) and may thus play
et al., 2000). The finding that drugs that inhibited either a role in depression. The three MAP kinase pathways, (1)
CRH1 or arginine vasopressin (AVP)1b receptors pre- extracellular signal-regulated kinase (ERK), (2) c-Jun N-
vented the reduced neurogenesis associated with a terminal kinase (JNK; also know as stress-activated pro-
chronic mild stressor (Alonso et al., 2004) points to an tein kinase) and (3) p38, are thought to play an important
important role for these neuropeptides in the hippocampal role in cellular responses to environmental events through
changes evident in affective illness. the transmission of synaptic signals to the nucleus (Curtis
and Finkbeiner, 1999). These pathways comprise a series
Changes of neuroplasticity in depression. Indirect of enzymes that sequentially phosphorylate one another to
evidence has pointed to the possibility that apoptotic pro- promote transcriptional activation and synthesis of proteins
cesses might be operative in depression. Specifically, important for cellular survival/death as well as inflamma-
postmortem analysis revealed modest, in situ end-labeling tory processes.
for DNA fragmentation in 11 of 15 depressed cases (Lu- Consistent with a role for MAP kinase pathways in
cassen et al., 2001). However, data are unavailable con- depression, it was shown that olfactory bulbectomy (which
cerning the expression of the prototypical apoptotic initia- elicits depressive-like behaviors in rodents) provoked long-
tors, Fas, p53, Bax and downstream caspases in clinical term expression of the immediate early gene, c-Jun, within
depression. Importantly, however, it will be recalled that the amygdala (Wrynn et al., 2000). c-Jun acts as a final
animal studies indicated that stressor exposure reduced messenger for the JNK pathway and controls the expres-
expression of the anti-apoptotic factor, bcl-2 (Manji and sion of several genes that promote either adaptive neuro-
Chen, 2002; Manji et al., 2003), and exposure to a severe plastic changes to environmental challenges (e.g. thermal
stressor exacerbated infarct size in response to ischemia or osmotic stress) or apoptotic responses to highly toxic
through the suppression of bcl-2 expression (Manji et al., insults, including trophic factor withdrawal, traumatic brain
2003). injury or neurotoxin exposure (Cowan and Storey, 2003;
Reductions of BDNF and bcl-2 expression elicited by Crocker et al., 2001; Hayley et al., 2004b; Saporito et al.,
stressors have been linked to altered levels of neurotrophic 2002). Rodents subjected to electroconvulsive shock,
factors and induction of mitogen-activated protein (MAP) which is used in the treatment of depression, displayed
kinase pathways (Manji and Chen, 2002). In this respect, it augmented JNK expression within the amygdala and hip-
was shown that BDNF was reduced in the serum of de- pocampus (Brecht et al., 1999). In these same animals,
pressed patients and the reduction correlated negatively this treatment initially enhanced phosphorylation (activa-
with the degree of clinical impairment (Shimizu et al., tion) of c-Jun within the amygdala and hypothalamus, but
2003). Moreover, several clinically beneficial treatments, diminished with progressive treatment (Brecht et al.,
including selective serotonin reuptake inhibitors (SSRIs), 1999), possibly reflecting ongoing neuroplastic changes
tricyclics and electroconvulsive therapy, increased BDNF corresponding to the clinical efficacy of electroconvulsive
expression (Shirayama et al., 2002). In rodents, it was therapy. As well, protein and mRNA for the MAP kinase,
similarly observed that both acute and chronic immobiliza- ERK, were reduced in the hippocampus and PFC of de-
tion reduced BDNF in the hippocampal dentate gyrus pressed suicide victims (Dwivedi et al., 2001). However,
(Smith et al., 1995), and a psychological stressor (explicit treatment with the antidepressant, fluoxetine, provoked
or contextual cues previously paired with footshock) like- expression of the neurotrophic factors, glial-derived neu-
wise reduced BDNF mRNA (Rasmusson et al., 2002). rotrophic factor (GDNF) and deiodinase 3, through activa-
Inasmuch as the reduced BDNF associated with stressors tion of the ERK and p38 kinase pathways, whereas BDNF
was not evident in adrenalectomized animals, the reduced expression was dependent on alternate signaling path-
levels of the neurotrophin were not attributable to stressor- ways (Mercier et al., 2004). Thus, the possibility exists that
666 S. Hayley et al. / Neuroscience 135 (2005) 659 – 678

MAP kinases may serve as an interface between signals that the CREB, BDNF, 5-HT and CRH differences between
regulating neuronal survival and plasticity and those asso- controls and depressed suicides that have been reported
ciated with emotional state. by others reflect post-transcriptional effects.
In accordance with the position that MAP kinases may Paralleling some of the findings in human studies,
influence or interact with cytokines, and conversely cyto- repeated exposure to inescapable footshock that elicited
kines may affect MAP kinases, it was reported that the p38 behavioral impairments in rats tested in the learned help-
MAP kinase pathway might be responsible for the central lessness model of depression, was associated with dimin-
induction of tumor necrosis factor-␣ (TNF-␣) and interleu- ished [(3)H]cAMP binding, PKA activity, and expression of
kin-1 (IL-1)␤ following traumatic ischemic injury (Piao et PKA RIIbeta and Calpha and Cbeta subunits, much like
al., 2003). In peripheral tissues, it was likewise observed those observed in the suicide brains (Dwivedi et al.,
that in response to infection (Mycobacterium avium) mac- 2004b). Further, overexpression of CREB within the amyg-
rophages produce high levels of TNF-␣ through p38 and dala, which may be important for anxiety associated with
CREB signaling (Roach et al., 2005). Conversely, several depression, was associated with an antidepressant-like
studies also indicated that IL-1␤ influences central expres- response in the learned helplessness model of depression
sion of p38, ERK, and JNK central signaling (Lai et al., (Wallace et al., 2004). Importantly, as well, the reduced
2003; Maher et al., 2004; Minogue et al., 2003). Likewise, CREB expression reported in rodents exposed to a chronic
some of the effects of IL-1, including long term potentia- stressor was up-regulated by 5-HT acting antidepressant
tion, as well as the neurodegenerative effects of the cyto- treatments (Lai et al., 2003; Shen et al., 2004). Thus, in our
kine, appear to be linked to alterations of JNK and ERK scheme, CREB activation may arise through increased
signaling pathways (Minogue et al., 2003; Wang et al., 5-HT activity (Fig. 1) that would facilitate BDNF production
2004). and ultimately modulate plasticity and hence positive mood
In addition to MAP kinases, the transcription factor, change.
cAMP response element binding protein (CREB) may con-
tribute to the regulation of mood state and neuroplasticity Neurotrophins and cytokines
(D’Sa and Duman, 2002). Evidence for the role of CREB in Cytokines, such as IL-1, IL-2, IL-6 and TNF-␣, serve as
hippocampal plasticity comes from the finding that CREB growth factors in peripheral immune cells. These signaling
activity, which is regulated upstream by PKA, modulated molecules and their receptors have also been identified
hippocampal long-term potentiation (Nguyen and Woo, within the brain, increase in response to traumatic brain
2003). Moreover, endogenous PKA activity was reduced in injury and neurological disturbances (Roth et al., 2004;
the PFC of depressed suicide victims, particularly with Rothwell, 1999; Vezzani et al., 2000; Wang and Shuaib,
respect to the RIIbeta and the catalytic Cbeta regulatory 2002), and may contribute to depressive states (Maes,
subunits (Dwivedi et al., 2004a). Likewise, in the dorsome- 1999). Depending on a number of factors (e.g. cytokine
dial PFC (granular frontal) of depressed suicides, mRNA concentrations, timing of application, endogenous vs ex-
and protein levels of CREB as well as PKA were dimin- ogenous source), cytokines may act either in a reparatory
ished (Dwivedi et al., 2003a,b). Postmortem analysis of capacity or promote neuronal damage (Stoll et al., 2000;
antidepressant drug-free patients that had suffered from Allan and Rothwell, 2001). We suggest that the proinflam-
major depression also revealed reduced levels of CREB in matory cytokines, IL-1, IL-6, TNF-␣ and IFN-␣/␥ promote
both its basal and activated (phosphorylated) state in the deleterious neuronal consequences through activation of
orbitofrontal cortex (Yamada et al., 2003). Further, al- neuroinflammatory cascades involving microglial release
though limited information is available concerning the in- of oxidative species. As depicted in Fig. 1, stressors may
fluence of antidepressants on CREB within brain, success- directly or indirectly (through HPA activity) come to af-
ful antidepressant-induced clinical responses were asso- fect cytokine functioning, thereby influencing depression
ciated with increased CREB phosphorylation (activation) in through several downstream processes.
peripheral T lymphocytes (Koch et al., 2002). Psychogenic insults affect circulating cytokine levels,
In contrast to these findings, increased cortical levels which may influence the organism’s ability to respond to
of CREB have also been reported in suicide victims; how- inflammatory or immunological challenges. The nature of
ever, these comprised a mixed group of major depressive the stressor effects (i.e. immunoenhancement or immuno-
and bipolar disorder patients (Odagaki et al., 2001) and as disturbance) depends on factors such as stressor chronic-
already indicated, subtypes of the disorder may involve ity (Dhabhar, 2000), and it seems that stressors may pref-
different signaling mechanisms. In a more recent and de- erentially affect certain cytokines (Zhang et al., 1998). It is
tailed analysis, Sibille et al. (2004) observed that within the of particular relevance to the present argument that just as
dosrsomedial and orbital PFC, neither CREB nor BDNF acute stressors influence peripheral cytokine activity, such
gene expression differed between controls and depressed challenges may also increase cytokine mRNA expression
suicides. As described earlier with respect to CRH and in brain and increase cytokine protein levels (Miyahara et
5-HT, the possibility exists that the absence of gene ex- al., 2000; Nguyen et al., 1998). In this regard, it was
pression differences may have been limited to these brain reported that a neurogenic stressor increased levels of IL-1
regions, or the genomic assays were not sufficiently sen- in the hypothalamus (Shintani et al., 1995) and in the
sitive to detect subtle differences that may exist. Of course, hippocampus and nucleus tractus solitarius (Nguyen et al.,
it is also conceivable, as indicated by Sibille et al. (2004), 1998). Moreover, pretreatment with the interleukin-1 re-
S. Hayley et al. / Neuroscience 135 (2005) 659 – 678 667

ceptor antagonist (IL-1ra) prevented the hormonal and cytokines are capable of eliciting depressive-like states.
hypothalamic monoamine alterations otherwise elicited by Indeed, systemic cytokine challenges may induce many of
a psychogenic stressor (Shintani et al., 1995). Together, the neurochemical alterations ordinarily associated with
these findings raise the possibility that the effects of stres- processive stressors (i.e. psychogenic or neurogenic
sors on central monoamine activity may be mediated stressors that involve appraisal of a situation, thus entailing
through central cytokine actions. higher order cortical processing) (Anisman and Merali,
Ultimately, neurotrophic factors such as BDNF may be 1999; Anisman et al., 2003). In particular, like psychogenic
affected by cytokines (possibly through their actions on stressors, IL-1, IL-6 and TNF-␣ provoke a constellation of
5-HT, see Fig. 1), and conversely, certain brain cytokines behavioral, neuroendocrine and central neurotransmitter
(e.g. IL-1␤) may be influenced by BDNF. Moreover, primed changes that have been implicated in depression. These
T helper lymphocytes that release anti-inflammatory cyto- include increased plasma levels of glucocorticoids and
kines, including IL-4 and IL-10, also induce BDNF expres- ACTH, coupled with augmented turnover of monoamines
sion (Ziemssen et al., 2002). Moreover, intra-hippocampal within hypothalamic and extrahypothalamic regions (Anis-
infusion of an IL-1 antagonist prevented the reduction of man et al., 2003; Hayley et al., 2003). Moreover, as de-
BDNF mRNA within the dentate gyrus and CA3 region scribed earlier, interplay may exist between neurotrophic
following social isolation. (Barrientos et al., 2004). In light factors and cytokines, which may affect depressive illness
of such findings it ought to be considered that cytokines either through the neurochemical changes imparted or by
may influence mood states as a result of processes related affecting neurogenesis and cell survival (Fig. 1).
to neuroplasticity, just as interplay between cytokines and
neurotrophins has been implicated in neurodegenerative Cytokine effects on neurochemical activity. Like psy-
illnesses, such as Parkinson’s (Nagatsu et al., 2000). In- chogenic and neurogenic stressors, systemic IL-1␤ in-
deed, it may well be that the reported high comorbidity creased 5-HT utilization within the paraventricular nucleus
between depression and Parkinson’s disease (McDonald of the hypothalamus (PVN), central amygdala, and mPFC.
et al., 2003), may involve similar processes, namely acti- Moreover, in vivo studies indicated that IL-1␤ administered
vation of proinflammatory cytokines and/or inhibition of systemically or directly into the brain increased hypotha-
anti-inflammatory factors (McDonald et al., 2003). The lamic NE release as well as that of 5-HT at several limbic
same holds true of the high comorbidity that exists be- sites (Kamikawa et al., 1998; Linthorst et al., 1995; Merali
tween depression and heart disease (particularly arthero- et al., 1997; Shintani et al., 1993; Song et al., 1999). The
sclerosis) where high levels of proinflammatory cytokines data concerning the effects of TNF-␣ are less extensive
have been detected along with c-reactive protein (Frasure- than those regarding IL-1, but its systemic administration
Smith et al., 2002). increased NE and 5-HT utilization in several brain regions
Certain cytokines, such as IL-6, are considered neuro- thought to contribute to depression and anxiety (Ando and
trophic factors as they play a significant role in the preven- Dunn, 1999; Brebner et al., 2000; Hayley et al., 1999).
tion of apoptosis and in the differentiation and regeneration In addition to the similarities between the effects of
within the CNS (Obara and Nakahata, 2002). Alternatively, stressors and cytokines, these treatments may synergisti-
cytokines may have neurotrophic actions through their cally increase monoamine release from limbic neurons
effects on BDNF (Murphy et al., 2000). Within peripheral (Merali et al., 1997; Song et al., 1999). One can likewise
blood mononuclear cells BDNF also influenced mRNA imagine, that the impact of cytokine elevations in humans
expression of TNF-␣, IL-4, as well as transforming growth might be influenced by the presence of ongoing life stres-
factor-␤ (Bayas et al., 2003). Whether such interactions sors or by subsyndromal levels of depression. As such,
also occur in brain is uncertain, although it was reported analyses of the relationship between inflammatory pro-
that systemic lipopolysaccharide (LPS) administration, at cesses and depressive state may benefit from assessing
doses that potently increase cytokine activity did not influ- the moderating role of psychosocial factors.
ence BDNF expression within the dentate gyrus (Shaw et While our position has been that the neurochemical
al., 2001). Yet, it will be recalled that intrahippocampal alterations imparted by cytokines may influence affective
pretreatment with IL-1ra prevented the reduction of BDNF state (Anisman and Merali, 1999), we emphasize again
in the CA1, CA2 and dentate gyrus otherwise elicited by that this should not be interpreted to suggest that the
social isolation (Barrientos et al., 2003). Together, these effects of systemic and psychogenic stressors are identi-
data are consistent with the view that cytokines may act in cal, nor does it imply that they will have comparable emo-
concert with BDNF in affecting cognitive or emotional tional or cognitive repercussions. It would certainly not be
processes. expected that systemic insults, such as immune chal-
lenges, would provoke cognitive disturbances akin to those
Stress– cytokine– depression connection
elicited by psychogenic challenges, although the influence
As indicated earlier, through their effects on neuroendo- of systemic insults may be sensitive to contextual or ex-
crine and neurotransmitter functioning, stressors may be periential (e.g. stressor) factors. Furthermore, cytokines
fundamental in the provocation of affective disorders. may stimulate HPA activity and other central functions
Thus, in relating depression to cytokines or to growth through pathways different from those utilized by psycho-
factors, it would seem propitious to consider whether stres- genic (e.g. predator exposure, restraint) or neurogenic
sors affect these physiological messengers, and whether (e.g. painful stimuli) stressors (Buller et al., 2001; Dayas et
668 S. Hayley et al. / Neuroscience 135 (2005) 659 – 678

al., 1999, 2001; Herman and Cullinan, 1997). Thus, just as tributed to the stressor resilience seen in adulthood (Liu et
multiple pathways may exist by which stressors come to al., 2000).
affect mood state (Fig. 1), different stressors may activate Just as limited maternal attention (extended separation
diverse neural circuits, and hence may not be equally from the dam or low levels of licking and grooming by the
effective in promoting depressive symptoms. dam) favored elevated neuroendocrine responses to stres-
At this juncture, it is essential to underscore that the sors encountered in adulthood (Meaney, 2001), rat pups
bulk of the available data have been limited to the effects exposed to a bacterial endotoxin, subsequently (as adults)
of acute cytokine/immune manipulations and limited infor- exhibited increased stressor-elicited corticosterone and
mation is available concerning the influence of chronic neuroimmune responses (Shanks et al., 2000; Boisse et
activation of the inflammatory immune system. Among al., 2004). Whether this neuroendocrine plasticity involves
other things, chronic cytokine elevations (as in the case of the same systems as those governing the stress sensiti-
IL-2) may result in variations of blood– brain barrier perme- zation is uncertain, but it does seem to involve at least
ability (Banks et al., 2004), thus permitting cytokines (and some of the same circuitry. Of particular relevance to the
present report is that early life immunological insults, like
other large molecules) greater accessibility to brain sites.
other stressors, could potentially affect vulnerability to de-
Furthermore, as in the case of stressors (Anisman and
pression owing to the greater reactivity of stress-relevant
Matheson, 2004), it may be more relevant to assess the
circuits, particularly those related to CRH and monoamine
effects of chronic cytokine elevations or repeated cytokine
activity.
treatments on depressive symptoms, as viral and bacterial
It is uncertain whether sensitization processes associ-
encounters typically occur on a chronic or subchronic ba- ated with stressors or cytokines affect BDNF, bcl-2 or other
sis. Under such conditions, the excessive wear and tear on normally protective factors, although there are inferential
physiological systems may be sufficiently great (allostatic data consistent with this possibility. Specifically, adminis-
overload), thus rendering the organism less well prepared tration of BDNF into the ventral tegmentum may elicit a
to contend with further insults (McEwen, 2001). behavioral sensitization characterized by progressively
Proactive effects of cytokines and stressors. Sys- greater motor activity with repeated treatments (Pierce et
temic and central cytokine administration increases the al., 1999; Guillin et al., 2001). As well, BDNF induced a
response to stressors or further cytokine exposure (Anis- behavioral sensitization to L-DOPA by triggering overex-
man et al., 2003; Hayley et al., 2003; Schmidt et al., 1996; pression of the D3 receptor within the basal ganglia (Guillin
et al., 2001; Sokoloff et al., 2002).
Tilders and Schmidt, 1999; Tilders et al., 1993). For in-
stance, stressors not only increased central IL-1␤ levels, Cytokines in depression
but also augmented the effects of subsequent central ad-
ministration of LPS on brain IL-1 levels. In contrast, among Animal models: sickness and depression. Adminis-
rats pretreated with IL-1ra the effects of the stressor treat- tration of cytokines, such as IL-1␤, or activation of macro-
ment were attenuated (Johnson et al., 2004). In addition to phages and other inflammatory immune cells by systemic
the sensitization of cytokine processes in brain, stressors, LPS treatment, provokes behavioral symptoms (reduced
IL-1␤ and TNF-␣ time-dependently increased the co-ex- locomotion, increased sleep, curled body posture, ptosis,
pression of CRH and AVP within the external zone of the piloerection) collectively referred to as sickness behavior
median eminence, although the time course varied across (Maier and Watkins, 1998; Dantzer, 2001). Although these
treatments. Thus, the co-release of CRH and AVP upon behaviors are thought to be of adaptive significance, as
they may serve to minimize energy expenditure, they are
subsequent challenge with stressor or cytokine treatments
also reminiscent of the neurovegetative features of depres-
synergistically stimulated ACTH secretion from the anterior
sion, albeit in themselves they do not reflect the core
pituitary (Hayley et al., 1999, 2002; Schmidt et al., 1996;
symptoms of depression. However, in addition to the sick-
Tilders and Schmidt, 1999). On the basis of such findings
ness behaviors, cytokines disrupt operant responding
the possibility exists that exposure to stressful events (in-
for reinforcement and elicit reduced social exploration
cluding activation of the inflammatory immune system)
(Dantzer, 2001; Merali et al., 2003). Although such effects
may dispose animals to greater vulnerability to stressor- may be secondary to the malaise induced by cytokines,
related pathology. The time course for these vulnerabilities rather than characteristics of depression per se, these
is not fully known, nor is it clear whether there are periods same features may reflect changes in motivational state
during which animals are particularly vulnerable to the (e.g. anhedonia). In fact, studies employing progressive
development of sensitized responses. However, numerous ratio schedules of reinforcement (i.e. these tap into the
reports have indicated that adverse events encountered degree to which an animal is willing to respond in order to
early in life may augment the adult response to stressors, gain a reward) indicated that cytokines act on incentive
whereas positive events (e.g. high levels of maternal care) motivational forces (drive) independent of effects related to
may have the effect of buffering against the effects of later malaise (Larson et al., 2002; Merali et al., 2003). More-
stressors (Meaney, 2001). While any number of factors over, antidepressant treatment attenuated the decreased
could potentially influence these processes, it is significant responding for a palatable snack ordinarily elicited by
that early life stimulation related to maternal care in- IL-1␤, but without influencing the reduced chow intake
creased BDNF mRNA expression and may thus have con- provoked by the cytokine (Merali et al., 2003).
S. Hayley et al. / Neuroscience 135 (2005) 659 – 678 669

As in the case of food-motivated behavior, it has been agnosed as reflecting major depression, sometimes of
shown that LPS may affect responding for rewarding brain sufficient severity to necessitate discontinuation of therapy
stimulation (in this instance electrodes were positioned (Denicoff et al., 1987; Meyers and Valentine, 1995;
within the lateral hypothalamus) (Borowski et al., 1998). As Miyaoka et al., 1999; Musselman et al., 2001; Maes et al.,
this paradigm does not involve food reward, the reduced 2001; Bonaccorso et al., 2002a,b; Capuron et al., 2000,
responding cannot be ascribed to anorexia, but instead 2002; Wichers and Maes, 2002; Maes and Bonaccorso,
likely reflects disturbances of incentive motivation. How- 2004). Not surprisingly, patients that exhibited subsyndro-
ever, it did appear that the effectiveness of LPS in disrupt- mal levels of depression prior to cytokine therapy were
ing responding was related to the intensity of the stimula- those most likely to develop the most pronounced depres-
tion, possibly suggesting that the effects of the treatment sive illness in response to the cytokine (Capuron et al.,
reflected the cost:benefit relationship. Together, these 2004).
data, and other related findings have supported the view It is of both practical and theoretical importance that
that beyond the neurovegetative effects of inflammatory depressive symptoms elicited by IFN-␣ were attenuated
immune activation, the treatments may promote anhedo- among patients that received conventional SSRI antide-
nia, a fundamental feature of depressive illness. pressant treatments, including sertraline, citalopram and
paroxetine (Schramm et al., 2000; Musselman et al., 2001;
Clinical evidence for cytokines in depression. De- Hauser et al., 2002; Kraus et al., 2002). Moreover, the
pressed patients, particularly those presenting with melan- course of symptom evolution, as well as the amelioration of
cholic illness, exhibit disturbances of several aspects of symptoms in relation to antidepressant medication, sug-
immune functioning, including altered circulating lympho- gests that the neurovegetative and mood-related symp-
cyte subsets, reduced mitogen-stimulated lymphocyte pro- toms introduced by immunotherapy are independent of
liferation, and impaired natural killer cytotoxicity, although one another. Specifically, the neurovegetative symptoms
equivocal results have been reported (Irwin, 1999; Maes, elicited by IFN-␣ (e.g. anorexia, fatigue and pain) fre-
1999). Yet, depression was also associated with immune quently appeared within 2 weeks of treatment commence-
activation reminiscent of an acute phase response, includ- ment, whereas depressed mood, anxiety and cognitive
ing increased plasma concentrations of complement pro- dysfunction tended to appear later. The latter symptoms
teins, C3 and C4, IgM, and positive acute phase proteins, were also more responsive to antidepressant medication
haptoglobin, ␣1-antitrypsin, ␣1 and ␣2 macroglobulin, and than were the neurovegetative symptoms (Capuron et
reduced negative acute phase proteins (Maes, 1999; Ro- al., 2002). These findings raise the possibility that the
thermundt et al., 2001). cytokine effects involve more than a single process, and
Commensurate with the view that depressive illness as such it is reasonable to expect that depression
might be secondary to activation of the inflammatory im- to systemic challenges that activate the inflammatory
mune system (Maes, 1999), severe depressive illness was immune system might likewise induce disparate symp-
accompanied by elevated circulating cytokines or their toms.
soluble receptors, including IL-2, soluble IL-2 receptors As described earlier, cytokine-elicited depression may
(sIL-2R), IL-1␤, IL-1ra, IL-6, soluble IL-6 receptor (sIL-6R), evolve owing to effects on biogenic amines, or by their
and ␥-interferon (IFN) (Anisman et al., 1999; Maes, 1999; potential impact on BDNF and bcl-2 expression, thereby
Mullar and Ackenheil, 1998; Nassberger and Traskman- affecting cellular viability and hence functioning. However,
Bendz, 1993; Sluzewska, 1999). Moreover, increased pro- Maes et al. (2001), Capuron et al., (2003) and Bonaccorso
duction of IL-1␤, IL-6 and TNF-␣ was evident in mitogen- et al. (2002a,b) provided an alternative scenario as to how
stimulated lymphocytes, and the magnitude of these effects cytokines might influence neurochemical functioning, while
was related to severity of illness (Maes, 1999), and chronicity taking into account the effects on cell survival. Specifically,
of illness in the case of IL-1␤ (Anisman et al., 1999). as illustrated in Fig. 1, these investigators indicated that
In addition to cytokine differences between depressed IFN-␣ as well as several proinflammatory cytokines
and non-depressed individuals, cytokines or immune chal- (TNF-␣, IL-1␤, IFN-␥) increased the activity of the 5-HT
lenges in humans may provoke depressive-like symptom- transporter, which may reduce extracellular 5-HT levels.
atology. For instance, healthy volunteers that received a As will be recalled, IFN-␣ increases indoleamine 2,3-dioxy-
low dose of LPS or vaccinated with live attenuated rubella genase (IDO) which is responsible for the catabolism of
virus displayed depressed mood up to 10 weeks following tryptophan to kynurenine (Menkes and MacDonald, 2000).
the challenge, and protracted periods of anxiety and mem- Thus, elevations of this enzyme may diminish the avail-
ory deficits were elicited by endotoxin (Yirmiya et al., ability of tryptophan and hence reduce brain 5-HT con-
1999). Furthermore, among patients undergoing IL-2 or centrations. In fact, depressive affect that followed cy-
IFN-␣ immunotherapy for hepatitis C or certain forms of tokine therapy was accompanied by reduced tryptophan
cancer, marked cognitive disturbances and neurovegeta- and elevated levels of kynurenine (Bonaccorso et al.,
tive symptoms frequently emerged (Valentine et al., 1995; 2002a,b). Yet, despite the inverse relationship between
Dieperink et al., 2000, 2003). Generally, these neuropsy- tryptophan and depressive symptoms, one cannot con-
chiatric symptoms included fatigue, sleep disturbances, clude a causal relationship from these data. Among
irritability, appetite suppression and depressed mood. In other things, metabolites of kynurenine, such as 3-hy-
fact, the affective symptoms that often emerged were di- droxy-kynurenine and quinolinic acid, through the induc-
670 S. Hayley et al. / Neuroscience 135 (2005) 659 – 678

tion of reactive oxygen species or stimulation of hip- disorders (e.g. Parkinsonism, Alzheimer’s disease, multi-
pocampal NMDA receptors, may promote tissue dam- ple sclerosis), and may thus contribute to cognitive and
age leading to depression (Wichers and Maes, 2004). mood processes associated with these illnesses (Pocock
Furthermore, as described in Fig. 1, it is possible that and Liddle, 2001; Hanisch, 2002; Vezzani et al., 2002).
cytokines, through their actions on 5-HT and BDNF
(which co-regulate one another) may contribute to the Concluding remarks
pathogenesis of depression as well as age-related neu- As depicted in Fig. 1, multiple factors may contribute to
rodegenerative disorders (Mattson et al., 2004). depression by impairing neuronal plasticity and disturbing
neurochemical functioning in key mood regulatory brain
Neurodegenerative aspects of depression: cytokine in-
regions. In this review we suggested that cytokines play an
volvement. Cytokines and other neuroinflammatory fac-
integral role in provoking these two outcomes through their
tors are in a position to influence cellular viability, and there
pleiotropic effects upon several physiological systems, in-
is a substantial literature exploring the potential effects of
cluding (1) HPA axis activity and CRH activity at limbic
cytokines on neurodegeneration. Certainly, this field has
sites, (2) monoamine utilization, (3) microglial release of
been controversial, and it will be recalled that some find-
oxidative species, (4) apoptotic pathways and (5) MAP
ings indicate that cytokines act in a reparatory capacity,
kinase and CREB signaling. Essentially, we argued that
whereas other studies suggest that cytokines function in a stressors and other environmental insults augment proin-
neurodestructive manner (Allan and Rothwell, 2001). flammatory cytokine expression while concomitantly low-
These differential effects may be related to cytokine con- ering levels of normally beneficial growth or neuroprotec-
centrations and genetic background of the host organism, tive factors. Such effects trigger a cascade of molecular
as well as timing of exposure to the cytokine relative to events involving promotion of inflammatory and apoptotic
various to insults. These differences not withstanding, con- pathways, which may ultimately impair cellular plasticity or
siderable evidence has indicated that proinflammatory cy- even survival and hence lead to abnormal neurotransmis-
tokines, notably TNF-␣ and IL-1␤, provoke neuronal loss sion favoring the evolution of depression.
through several possible mechanisms. In particular, cyto- To be sure, the processes that subserve stressor-
kines, such as TNF-␣, may induce apoptotic conse- provoked depression remain to be fully elucidated. How-
quences due to their actions on intracellular caspase-re- ever, evidence exists supporting a role for 5-HT and CRH
lated pathways (Varfolomeev and Ashkenazi, 2004), or involvement in depression, and that 5-HT variations may
they may affect cell survival through extracellular mecha- inhibit GABAA receptor subunit expression, thereby pro-
nisms, including excitotoxic processes that involve gluta- moting depressive symptoms. As well, through its action
mate activity (Silverstein et al., 1997; Rothwell, 1999; on ACTH and glucocorticoids, CRH may influence cytokine
Allan, 2002). Additionally, the proinflammatory cytokine, functioning and its downstream processes, including 5-HT
IFN-␥, as well as endotoxins may elicit the release of free changes as well as neuroplasticity. In addition to the path-
radicals from microglia (Pawate et al., 2004), thereby pro- ways depicted in the figure, corticoids may also result in
moting oxidative injury. Finally, as described earlier, cyto- increased deoxycorticosterone release and the production
kines might mediate neurodegeneration through the pro- of its metabolite, tetrahydroxydeoxycorticosterone (THDOC),
motion of metabolic toxins, such as 3-hydroxy-kynurenine which has a positive effect on GABAA functioning (Reddy,
and quinolinc acid, which themselves are increased in 2003).
depression (Wichers and Maes, 2004). These kynurenine Beyond the potential role for CRH and monoamine
metabolites can synergistically induce free radical gener- disturbances, variations of growth factors and anti-apop-
ation and have been implicated in a number of neurode- totic factors, as well as cytokines, might contribute to de-
generative diseases, including Huntington’s and Parkin- pression. Stressors, it will be recalled, reduce hippocampal
son’s, and have been implicated in AIDS dementia (Wich- BDNF and bcl-2, possibly through effects on corticoste-
ers and Maes, 2004). rone release, although other neural circuits may also lead
It will be recalled that macrophage-derived inflamma- to such an outcome. Stressors also increase cytokine ex-
tory factors were hypothesized to influence mood states pression in brain, which could promote CRH release,
(Smith, 1991). The common myeloid lineage and the strik- hence eliciting 5-HT neuronal alterations. Alternatively, cy-
ing similarity of functioning between peripheral macro- tokines may cause the reduction of 5-HT levels through
phages and brain microglia raise the possibility of common activation of indoleamine-pyrrole 2, 3-dioxygenase (IDO),
involvement of these cell types in depression. Indeed, the enzyme responsible for the catabolism of tryptophan to
microglia are the primary central reservoirs of proinflam- kynurenine. As depicted in Fig. 1, kynurenine metabolites
matory cytokines, and like macrophages, they act as anti- (3-hydroxy-kynurenine and quinolinic acid) affect reactive
gen presenting cells within the brain (Hanisch, 2002). oxygen species or stimulate hippocampal NMDA recep-
Thus, these cells may function to orchestrate central im- tors, hence promoting tissue damage, possibly leading to
mune responses that may have deleterious consequences depression (Wichers and Maes, 2004).
on local tissue. In this regard, microglia release several Ultimately, in considering the processes that subserve
cytokines (IL-1, IL-6, IL-8, TNF-␣ and IFN-␥) in response to depressive illness it is essential to account for some of the
various stressful and traumatic stimuli (e.g. seizure, stroke, phenomenological aspects of the disorder. Symptoms of
head injury) and over the course of neurodegenerative depression differ markedly across individuals (although
S. Hayley et al. / Neuroscience 135 (2005) 659 – 678 671

common features are typically present), and subtypes of individuals), interindividual and subtype differences may
depression exist that may involve different neurochemical exist with respect to the presence of certain neurochemical
substrates. Subtypes of depression are characterized by disturbances.
different symptom profiles, may be differentially responsive It will be recalled that depression is comorbid with a
to various classes of antidepressant therapy, and may variety of pathological states. These range from other psy-
involve different neurochemical substrates. For instance, chiatric disorders (e.g. posttraumatic stress disorder;
while typical depression involves reduced eating and Brady et al., 2000) or psychological symptoms (e.g. anxi-
sleep, in atypical depressive disorder reversed neuroveg- ety) through to heart disease (Frasure-Smith et al., 2002)
etative features predominate (i.e. increased eating, partic- and neurodegenerative disorders, such as Parkinsonism
ularly carbohydrate craving, increased sleep, fatigue; Gold (McDonald et al., 2003). It is unlikely that the psychological
and Chrousos, 2002). Likewise, melancholic depression distress associated with these illnesses provoked the de-
may involve processes in addition to those seen in less pression, as the affective disorder frequently preceded the
severe forms of depressive illness, and chronic depres- physical illness (e.g. Grippo et al., 2003a). Instead, it has
sion, even of moderate severity (dysthymia), may be as- been argued that the comorbidities stem from processes
sociated with biological processes that are less prominent common across disorders, and indeed each of these ill-
in more acute illness (Griffiths et al., 2000). Whereas mel- nesses can be linked to inflammatory processes (Maes,
ancholic depression may be related to CRH neuronal hy- 1999; Gao et al., 2003; Rattazzi et al., 2003; Grippo et al.,
perfunctioning, atypical depression was aligned with down- 2003a,b; Hayley et al., 2004). In effect, it is possible that
regulated HPA activity, including CRH deficiency (Anisman cytokines or related factors are a common denominator for
et al., 1999; Gold and Chrousos, 2002). Furthermore, as these diverse pathologies.
cytokine immunotherapy (using IFN-␣) gives rise to de- A third feature of depression that needs to be ad-
pression where neurovegetative symptoms appear in ad- dressed is the view that this disorder may be a life-long
vance of the affective features, and are more resistant to illness given its very high rate of recurrence (50 –70% in 5
antidepressant treatment than are the mood disturbances years; Keller, 2002). Coupled with this, is the fact that 5-HT
(Capuron et al., 2002), it might be considered that cytokine and NE acting antidepressants have positive treatment
activation has its primary effect on typical depression, effects (despite the high rates of placebo response) in only
while the neurovegetative changes are a reflection of sick- about two thirds of patients. To account for these aspects
ness provoked by the cytokine. of the disorder it could be argued that certain individuals
The elevated CRH associated with melancholic (typi- are more emotionally reactive to stressful events, or pos-
cal) depression is consistent with the potent HPA stimulat- sess a pre-morbid depressive personality, hence making
ing actions of pro-inflammatory cytokines. In a similar fash- them prone to depression given the presence of environ-
ion, the argument can be advanced that high levels of mental triggers. Alternatively, it can be argued that neuro-
comorbid anxiety may be related to elevated CRH func- nal and neurochemical changes akin to those described in
tioning in central amygdala or bed nucleus of the stria this review are responsible for the persistence of depres-
terminalis (Walker and Davis, 1997), or altered functioning sion. In particular, as indicated earlier, stressful experi-
of CRH and hence NE activity in locus coeruleus-related ences (including those encountered early in life) and acti-
processes (Asbach et al., 2001; Brady, 1994). Further- vation of the inflammatory immune system may result in
more, the presence of dopamine disturbances, particularly phenotypic changes of CRH neurons within the median
within tegmental and accumbal regions, may be linked to eminence, so that they come to co-express AVP (Tilders
depressive episodes where anhedonia is an essential fea- and Schmidt, 1999; Hayley et al., 2001). The conjoint
ture, especially as this transmitter has been aligned with release of these peptides may promote greater HPA acti-
reward processes (Wise, 2004). Likewise, given the role of vation and may thus contribute to depression. Importantly,
frontal cortical 5-HT in cognitive processes, behavioral the CRH/AVP co-expression is fairly long lasting and
planning (Kalivas and Nakamura, 1999; Tanji and Hoshi, hence may influence mood changes long after the initial
2001) and impulsive behaviors (Evenden, 1999), it is pos- stressor experience (Tilders and Schmidt, 1999). In fact, it
sible that disturbances of this transmitter might be associ- has been suggested that certain illnesses, such as dyst-
ated with suicide (Purselle and Nemeroff, 2003). One can hymia (low grade, chronic depression), are associated with
be less certain about the specific symptoms associated a change of basal CRH/AVP levels so that these individ-
with BDNF and bcl-2 variations, as most of the available uals are particularly vulnerable to major depressive illness
data have been obtained from studies of the hippocampus, (i.e. double depression) (Griffiths et al., 2000).
but there is reason to suppose that growth factors are Yet another alternative to accommodate the persis-
affected in other brain regions (e.g. animal studies re- tence of the depressive vulnerability concerns potential
vealed stressor effects on BDNF in cortical regions; Hashi- effects of stressors, as well as cytokines, on neuroplastic-
moto et al., 2004) and could potentially influence a variety ity. As already indicated, such insults, by virtue of effects
of neurotransmitter systems. Certainly, some of the sug- on BDNF and bcl-2, or by affecting the kyneurenine path-
gestions offered here are highly provisional. Nevertheless, way or other oxidative/excitotoxic processes, may result in
the array of neurochemical processes linked to depressive altered dendritic branching or suppression of neurogen-
illness suggests that while common underlying processes esis. From this perspective, antidepressants may be af-
may be associated with all types of depression (and in all fecting depressive states by altering factors responsible for
672 S. Hayley et al. / Neuroscience 135 (2005) 659 – 678

neuroplasticity (e.g. 5-HT functioning) without actually af- Asbach S, Schulz C, Lehnert H (2001) Effects of corticotropin-releas-
fecting those factors that lead to the 5-HT alterations (e.g. ing hormone on locus coeruleus neurons in vivo: a microdialysis
CRH/AVP), and hence may be masking symptoms rather study using a novel bilateral approach. Eur J Endocrinol 145:
359 –363.
than treating the disease. Thus, with treatment cessation,
Austin MC, Janosky JE, Murphy HA (2003) Increased corticotropin-
and the persistence of the initial neurochemical dysfunc- releasing hormone immunoreactivity in monoamine-containing
tion, major depression has an increased likelihood of re- pontine nuclei of depressed suicide men. Mol Psychiatry 8:324 –
emerging. Ultimately, it may be profitable to focus on com- 332.
bined therapeutic strategies that involve factors (e.g. cyto- Banki CM, Karmacsi L, Bissette G, Nemeroff CB (1992) CSF cortico-
kines) that come to affect neuroplasticity, or those that tropin-releasing hormone and somatostatin in major depression:
influence the way in which stressors come to affect neu- response to antidepressant treatment and relapse. Eur Neuropsy-
chopharmacol 2:107–113.
rotransmitter or neurohormone functioning.
Banks WA, Niehoff ML, Zalcman SS (2004) Permeability of the mouse
blood-brain barrier to murine interleukin-2: predominance of a sat-
Acknowledgments—Supported by funds from the Natural Science urable efflux system. Brain Behav Immun 18:434 – 442.
and Engineering Research Council of Canada (NSERC) and by Barrientos RM, Sprunger DB, Campeau S, Watkins LR, Rudy JW,
the Canadian Institutes of Health Research (CIHR). S.H. and H.A. Maier SF (2004) BDNF mRNA expression in rat hippocampus
are CIHR Canada Research Chairs. following contextual learning is blocked by intrahippocampal
IL-1beta administration. J Neuroimmunol 155:119 –126.
Barrientos RM, Sprunger DB, Campeau S, Higgins EA, Watkins LR,
REFERENCES Rudy JW, Maier SF (2003) Brain-derived neurotrophic factor
mRNA downregulation produced by social isolation is blocked by
Aguilera G, Rabadan-Diehl C, Nikodemova M (2001) Regulation of intrahippocampal interleukin-1 receptor antagonist. Neuroscience
pituitary corticotropin releasing hormone receptors. Peptides 22: 121:847– 853.
769 –774. Bayas A, Kruse N, Moriabadi NF, Weber F, Hummel V, Wohleben G,
Allan SM, Rothwell NJ (2001) Cytokines and acute neurodegenera- Gold R, Toyka KV, Rieckmann P (2003) Modulation of cytokine
tion. Nat Rev Neurosci 2:734 –744. mRNA expression by brain-derived neurotrophic factor and nerve
Allan SM (2002) Varied actions of proinflammatory cytokines on exci- growth factor in human immune cells. Neurosci Lett 335:155–158.
totoxic cell death in the rat central nervous system. J Neurosci Res Bissette G, Klimek V, Pan J, Stockmeier C, Ordway G (2003) Elevated
67:428 – 434. concentrations of CRF in the locus coeruleus of depressed sub-
Alonso R, Griebel G, Pavone G, Stemmelin J, Le Fur G, Soubrie P jects. Neuropsychopharmacology 28:1328 –1335.
(2004) Blockade of CRF(1) or V(1b) receptors reverses stress- Blier P (2003) The pharmacology of putative early-onset antidepres-
induced suppression of neurogenesis in a mouse model of depres- sant strategies. Eur Neuropsychopharmacol 13:57– 66.
sion. Mol Psychiatry 9:278 –286.
Boisse L, Mouihate A, Ellis S, Pittman QJ (2004) Long-term alterations
Altar CA (1999) Neurotrophins and depression. Increased cerebrospi-
in neuroimmune responses after neonatal exposure to lipopolysac-
nal fluid levels of neurotrophin 3 (NT-3) in elderly patients with
charide. J Neurosci 24:4928 – 4934.
major depression. Trends Pharmacol Sci 20:59 – 61.
Bonaccorso S, Marino V, Biondi M, Grimaldi F, Ippolit F, Maes M
Ando T, Dunn AJ (1999) Mouse tumor necrosis factor-alpha increases
(2002a) Depression induced by treatment with interferon-alpha in
brain tryptophan concentrations and norepinephrine metabolism
patients affected by hepatitis C virus. J Affect Disord 72:237–241.
while activating the HPA axis in mice. Neuroimmunomodulation
Bonaccorso S, Marino V, Puzella A, Pasquini M, Biondi M, Artini M,
6:319 –329.
Almerighi C, Verkerk R, Meltzer H, Maes M (2002b) Increased
Anguelova M, Benkelfat C, Turecki G (2003a) A systematic review of
depressive ratings in patients with hepatitis C receiving interferon-
association studies investigating genes coding for serotonin recep-
alpha-based immunotherapy are related to interferon-alpha-
tors and the serotonin transporter: I. Affective disorders. Mol Psy-
induced changes in the serotonergic system. J Clin Psychophar-
chiatry 8:574 –591.
macol 22:86 –90.
Anguelova M, Benkelfat C, Turecki G (2003b) A systematic review of
association studies investigating genes coding for serotonin recep- Borowski T, Kokkinidis L, Merali Z, Anisman H (1998) Lipopolysac-
tors and the serotonin transporter: II. Suicidal behavior. Mol Psy- charide, central in vivo biogenic amine variations, and anhedonia.
chiatry 8:646 – 653. Neuroreport 9:3797–3802.
Anisman H, Matheson K (2005) Anhedonia and depression: caveats Brady LS (1994) Stress, antidepressant drugs, and the locus coer-
concerning animal models. Neurosci Biobehav Rev 29, uleus. Brain Res Bull 35:545–556.
1525–1546. Brady KT, Killeen TK, Brewerton T, Lucerini S (2000) Comorbidity of
Anisman H, Merali Z, Hayley S (2003) Sensitization associated with psychiatric disorders and posttraumatic stress disorder. J Clin
stressors and cytokine treatments. Brain Behav Immun 17:86 –93. Psychiatry 7:22–32.
Anisman H, Merali Z (1999) Anhedonic and anxiogenic effects of Brambilla P, Perez J, Barale F, Schettini G, Soares JC (2003) GABAergic
cytokine exposure. Adv Exp Med Biol 461:199 –233. dysfunction in mood disorders. Mol Psychiatry 715:721–737.
Anisman H, Ravindran AV, Griffiths J, Merali Z (1999) Endocrine and Brebner K, Hayley S, Merali Z, Anisman H (2000) Synergistic effects of
cytokine correlates of major depression and dysthymia with typical interleukin-1, interleukin-6, and TNF-␣: neuroendocrine, neuro-
or atypical features. Mol Psychiatry 4:182–188. chemical and behavioral changes. Neuropsychopharmacology 22:
Arango V, Underwood MD, Gubbi AV, Mann JJ (1995) Localized 566 –580.
alterations in pre- and postsynaptic serotonin binding sites in the Brecht S, Simler S, Vergnes M, Mielke K, Marescaux C, Herdegen T
ventrolateral prefrontal cortex of suicide victims. Brain Res 688: (1999) Repetitive electroconvulsive seizures induce activity of
121–133. c-Jun N-terminal kinase and compartment-specific desensitization
Arato M, Banki CM, Bissette G, Nemeroff CB (1989) Elevated CSF of c-Jun phosphorylation in the rat brain. Mol Brain Res 68:
CRF in suicide victims. Biol Psychiatry 25:355–359. 101–108.
Arborelius L, Owens MJ, Plotsky PM, Nemeroff CB (1999) The role of Bremner JD, Narayan M, Anderson ER, Staib LH, Miller HL, Charney
corticotropin-releasing factor in depression and anxiety disorders. DS (2000) Hippocampal volume reduction in major depression.
J Endocrinol 160:1–12. Am J Psychiatry 157:115–118.
S. Hayley et al. / Neuroscience 135 (2005) 659 – 678 673

Brooks-Kayal AR, Shumate MD, Jin H, Lin DD, Rikhter TY, Holloway distinctive recruitment patterns in the amygdala and in medullary
KL, Coulter DA (1999) Human neuronal gamma-aminobutyric noradrenergic cell groups. Eur J Neurosci 14:1143–1152.
acid(A) receptors: coordinated subunit mRNA expression and Dayas CV, Buller KM, Day TA (1999) Neuroendocrine responses to an
functional correlates in individual dentate granule cells. J Neurosci emotional stressor: evidence for involvement of the medial but not
19:8312– 8318. the central amygdala. Eur J Neurosci 11:2312–2322.
Buller K, Xu Y, Dayas C, Day T (2001) Dorsal and ventral medullary Denicoff KD, Rubinow DR, Papa MZ, Simpson L, Seipp LA, Lotze MT,
catecholamine cell groups contribute differentially to systemic in- Chang AE, Rosenstein D, Rosenberg SA (1987) The neuropsychi-
terleukin-1beta-induced hypothalamic pituitary adrenal axis re- atric effects of treatment with interleukin-2 and lymphokine-acti-
sponses. Neuroendocrinology 73:129 –138. vated killer cells. Ann Intern Med 107:293–300.
Campbell S, Marriott M, Nahmias C, MacQueen GM (2004) Lower Dhabhar FS (2000) Acute stress enhances while chronic stress sup-
hippocampal volume in patients suffering from depression: a meta- presses skin immunity. The role of stress hormones and leukocyte
analysis. Am J Psychiatry 61:598 – 607. trafficking. Ann N Y Acad Sci 917:876 – 893.
Capuron L, Ravaud A, Miller AH, Dantzer R (2004) Baseline mood and Dieperink E, Ho SB, Thuras P, Willenbring ML (2003) A prospective
psychosocial characteristics of patients developing depressive study of neuropsychiatric symptoms associated with interferon-
symptoms during interleukin-2 and/or interferon-alpha cancer ther- alpha-2b and ribavirin therapy for patients with chronic hepatitis C.
apy. Brain Behav Immun 18:205–213. Psychosomatics 44:104 –112.
Capuron L, Raison CL, Musselman DL, Lawson DH, Nemeroff CB, Dieperink E, Willenbring M, Ho SB (2000) Neuropsychiatric symptoms
Miller AH (2003) Association of exaggerated HPA axis response to associated with hepatitis C and interferon alpha: A review. Am J
the initial injection of interferon-alpha with development of depres- Psychiatry 157:867– 876.
sion during interferon-alpha therapy. Am J Psychiatry 160: Drevets WC (2001) Neuroimaging and neuropathological studies of
1342–1345. depression: implications for the cognitive-emotional features of
Capuron L, Gumnick JF, Musselman DL, Lawson DH, Reemsnyder A, mood disorders. Curr Opin Neurobiol 11:240 –249.
Nemeroff CB, Miller AH (2002) Neurobehavioral effects of interfer- D’Sa C, Duman RS (2002) Antidepressants and neuroplasticity. Bipo-
on-alpha in cancer patients: phenomenology and paroxetine re- lar Disord 4:183–194.
sponsiveness of symptom dimensions. Neuropsychopharmacol- Du L, Faludi G, Palkovits M, Demeter E, Bakish D, Lapierre YD,
ogy 26:643– 652. Sotonyi P, Hrdina PD (1999) Frequency of long allele in serotonin
Capuron L, Ravaud A, Dantzer R (2000) Early depressive symptoms transporter gene is increased in depressed suicide victims. Biol
in cancer patients receiving interleukin 2 and/or interferon alfa-2b Psychiatry 46:196 –201.
therapy. J Clin Oncol 18:2143–2151. Duman RS (2004) Role of neurotrophic factors in the etiology and
Caspi A, Sugden K, Moffitt TE, Taylor A, Craig IW, Harrington H, treatment of mood disorders. Neuromol Med 5:11–25.
McClay J, Mill J, Martin J, Braithwaite A, Poulton R (2003) Influ- Duman RS, Malberg J, Thome J (1999) Neural plasticity to stress and
ence of life stress on depression: moderation by a polymorphism in antidepressant treatment. Biol Psychiatry 46:1181–1191.
the 5-HTT gene. Science 301:386 –389. Dwivedi Y, Rizavi HS, Shukla PK, Lyons J, Faludi G, Palkovits M,
Cheetham SC, Crompton MR, Katona CL, Parker SJ, Horton RW Sarosi A, Conley RR, Roberts RC, Tamminga CA, Pandey GN
(1988) Brain GABAA/benzodiazepine binding sites and glutamic (2004a) Protein kinase A in postmortem brain of depressed suicide
acid decarboxylase activity in depressed suicide victims. Brain Res victims: altered expression of specific regulatory and catalytic sub-
460:114 –123. units. Biol Psychiatry 55:234 –243.
Cook CJ (2001) Measuring of extracellular cortisol and corticotropin- Dwivedi Y, Mondal AC, Shukla PK, Rizavi HS, Lyons J (2004b) Altered
releasing hormone in the amygdala using immunosensor coupled protein kinase A in brain of learned helpless rats: effects of acute
microdialysis. J Neurosci Methods 110:95–101. and repeated stress. Biol Psychiatry 56:30 – 40.
Cook SC, Wellman CL (2004) Chronic stress alters dendritic morphol- Dwivedi Y, Rao JS, Rizavi HS, Kotowski J, Conley RR, Roberts RC,
ogy in rat medial prefrontal cortex. J Neurobiol 60:236 –248. Tamminga CA, Pandey GN (2003a) Abnormal expression and
Cowan KJ, Storey KB (2003) Mitogen-activated protein kinases: new functional characteristics of cyclic adenosine monophosphate re-
signaling pathways functioning in cellular responses to environ- sponse element binding protein in postmortem brain of suicide
mental stress. Exp Biol 206:1107–1115. subjects. Arch Gen Psychiatry 60:273–282.
Coyle JT, Duman RS (2003) Finding the intracellular signaling path- Dwivedi Y, Rizavi HS, Conley RR, Roberts RC, Tamminga CA, Pan-
ways affected by mood disorder treatments. Neuron 38:157–160. dey GN (2003b) Altered gene expression of brain-derived neuro-
Crocker SJ, Lamba WR, Smith PD, Callaghan SM, Slack RS, Anisman trophic factor and receptor tyrosine kinase B in postmortem brain of
H, Park DS (2001) c-Jun mediates axotomy-induced dopamine suicide subjects. Arch Gen Psychiatry 60:804 – 815.
neuron death in vivo. Proc Natl Acad Sci U S A 98:13385–13390. Dwivedi Y, Conley RR, Roberts RC, Tamminga CA, Pandey GN
Cross JA, Cheetham SC, Crompton MR, Katona CL, Horton RW (2002) [(3)H]cAMP binding sites and protein kinase a activity in the
(1988) Brain GABAB binding sites in depressed suicide victims. prefrontal cortex of suicide victims. Am J Psychiatry 159:66 –73.
Psychiatry Res 26:119 –129. Dwivedi Y, Rizavi HS, Roberts RC, Conley RC, Tamminga CA, Pan-
Cullinan WE, Wolfe TJ (2000) Chronic stress regulates levels of dey GN (2001) Reduced activation and expression of ERK1/2 MAP
mRNA transcripts encoding beta subunits of the GABA(A) receptor kinase in the post-mortem brain of depressed suicide subjects.
in the rat stress axis. Brain Res 887:118 –124. J Neurochem 77:916 –928.
Curtis J, Finkbeiner S (1999) Sending signals from the synapse to the Eom DS, Choi WS, Oh YJ (2004) Bcl-2 enhances neurite extension via
nucleus: possible roles for CaMK, Ras/ERK, and SAPK pathways activation of c-Jun N-terminal kinase. Biochem Biophys Res Comm
in the regulation of synaptic plasticity and neuronal growth. Neu- 314:377–381.
rosci Res 58:88 –95. Escriba PV, Ozaita A, Garcia-Sevilla JA (2004) Increased mRNA
Dantzer R (2001) Cytokine-induced sickness behavior: Where do we expression of alpha2A-adrenoceptors, serotonin receptors and
stand? Brain Behav Immun 15:7–24. mu-opioid receptors in the brains of suicide victims. Neuropsycho-
Dautzenberg FM, Hauger RL (2002) The CRF peptide family and their pharmacology 29:1512–1521.
receptors: yet more partners discovered. Trends Pharmacol Sci Evenden J (1999) Impulsivity: a discussion of clinical and experimental
23:71–77. findings. J Psychopharmacol 13:180 –192.
Dayas CV, Buller KM, Crane JW, Xu Y, Day TA (2001) Stressor Faludi G, Du L, Palkovits M, Antal B, Sotonyi P, Hrdina PD (2000)
categorization: acute physical and psychological stressors elicit Serotonin transporter, serotonin-2A receptor and tryptophan hy-
674 S. Hayley et al. / Neuroscience 135 (2005) 659 – 678

droxilase gene polymorphisms in depressed suicide victims. Hayley S, Staines WM, Merali Z, Anisman H (2001) Time-dependent
Neurobiology 8:269 –271. sensitization of corticotropin-releasing hormone, arginine vaso-
Frasure-Smith N, Lesperance F, Gravel G, Masson A, Juneau M, pressin and c-fos immunoreactivity within the mouse brain in re-
Bourassa MG (2002) Long-term survival differences among low- sponse to tumor necrosis factor-alpha. Neuroscience 106:
anxious, high-anxious and repressive copers enrolled in the Mon- 137–148.
treal heart attack readjustment trial. Psychosom Med 64:571–579. Hayley S, Wall P, Anisman H (2002) Sensitization to the neuroendo-
Frodl T, Meisenzahl EM, Zill P, Baghai T, Rujescu D, Leinsinger G, crine, central monoamine and behavioral effects of murine tumor
Bottlender R, Schule C, Zwanzger P, Engel RR, Rupprecht R, necrosis factor-␣: peripheral and central mechanisms. Eur J Neu-
Bondy B, Reiser M, Moller HJ (2004) Reduced hippocampal vol- rosci 15:1061–1076.
umes associated with the long variant of the serotonin transporter Hayley S, Anisman H (2004) Multiple mechanisms of cytokine action in
polymorphism in major depression. Arch Gen Psychiatry 61: neurodegenerative and psychiatric states: neurochemical and mo-
177–183. lecular substrates. Curr Pharm Design, in press.
Gao HM, Liu B, Zhang W, Hong JS (2003) Novel anti-inflammatory Hayley S, Crocker SJ, Smith PD, Shree T, Jackson-Lewis V, Przed-
therapy for Parkinson’s disease. Trends Pharmacol Sci 24: borski S, Mount M, Slack R, Anisman H, Park DS (2004b) Regu-
395– 401. lation of dopaminergic loss by Fas in a 1-methyl-4-phenyl-1,2,3,6-
Gilbertson MW, Shenton ME, Ciszewski A, Kasai K, Lasko NB, Orr SP, tetrahydropyridine model of Parkinson’s disease. J Neurosci 24:
Pitman RK (2002) Smaller hippocampal volume predicts patho- 2045–2053.
logic vulnerability to psychological trauma. Nat Neurosci 5: Hellsten J, Wennstrom M, Mohapel P, Ekdahl CT, Bengzon J, Ting-
1242–1247. strom A (2002)Electroconvulsive seizures increase hippocampal
Gilmor ML, Skelton KH, Nemeroff CB, Owens MJ (2003) The effects of neurogenesis after chronic corticosterone treatment. Eur J Neuro-
chronic treatment with the mood stabilizers valproic acid and lith- sci 16:283–290.
ium on corticotropin-releasing factor neuronal systems. J Pharma- Herman JP, Cullinan WE (1997) Neurocircuitry of stress: Central con-
col Exp Ther 305:434 – 439. trol of hypothalamo-pituitary-adrenocortical axis. Trends Neurosci
Givalois L, Arancibia S, Alonso G, Tapia-Arancibia L (2004) Expres- 20:78 – 84.
sion of brain-derived neurotrophic factor and its receptors in the Heuser I, Bissette G, Dettling M, Schweiger U, Gotthardt U, Schmider
median eminence cells with sensitivity to stress. Endocrinology J, Lammers CH, Nemeroff CB, Holsboer F (1998) Cerebrospinal
145:4737– 4747. fluid concentrations of corticotropin-releasing hormone, vasopres-
Gold PW, Chrousos GP (2002) Organization of the stress system and sin, and somatostatin in depressed patients and healthy controls:
its dysregulation in melancholic and atypical depression: high vs response to amitriptyline treatment. Depress Anxiety 8:71–79.
low CRH/NE states. Mol Psychiatry 7:254 –275. Hock C, Heese K, Muller-Spahn F, Huber P, Riesen W, Nitsch RM,
Gould E, McEwen BS (1993) Neuronal birth and death. Curr Opin Otten U (2000) Increased cerebrospinal fluid levels of neurotrophin
Neurobiol 3:676 – 682. 3 (NT-3) in elderly patients with major depression. Mol Psychiatry
Griffiths J, Ravindran AV, Merali Z, Anisman H (2000) Dysthymia: 5:510 –513.
Neurochemical and behavioral perspectives. Mol Psychiatry 5: Holsboer F (2003) Corticotropin-releasing hormone modulators and
242–261. depression. Curr Opin Investig Drugs 4:46 –50.
Grippo AJ, Beltz TG, Johnson AK (2003a) Behavioral and cardiovas- Honig A, Bartlett JR, Bouras N, Bridges PK (1988) Amino acid levels
cular changes in the chronic mild stress model of depression. in depression: a preliminary investigation. J Psychiatr Res 22:
Physiol Behav 78:703–710. 159 –164.
Grippo AJ, Francis J, Weiss RM, Felder RB, Johnson AK (2003b) Hucks D, Lowther S, Crompton MR, Katona CL, Horton RW (1997)
Cytokine mediation of experimental heart failure-induced anhedo- Corticotropin-releasing factor binding sites in cortex of depressed
nia. Am J Physiol Regul Integr Comp 284:R666 –R673. suicides. Psychopharmacology 199:174 –178.
Gross-Isseroff R, Biegon A, Voet H, Weizman A (1998) The suicide Iredale PA, Terwilliger R, Widnell KL, Nestler EJ, Duman RS (1996)
brain: a review of postmortem receptor/transporter binding studies. Differential regulation of corticotropin-releasing factor1 receptor
Neurosci Biobehav Rev 22:653– 661. expression by stress and agonist treatments in brain and cultured
Grossman R, Buchsbaum MS, Yehuda R (2002) Neuroimaging stud- cells. Mol Pharmacol 50:1103–1110.
ies in post-traumatic stress disorder. Psychiatr Clin North Am Irwin M (1999) Immune correlates of depression. Adv Exp Med Biol
25:317–340. 461:1–24.
Guillin O, Diaz J, Carroll P, Griffon N, Schwartz JC, Sokoloff P (2001) Jacobs BL (2002) Adult brain neurogenesis and depression. Brain
BDNF controls dopamine D3 receptor expression and triggers Behav Immun 16:602– 609.
behavioural sensitization. Nature 411:86 – 89. Johnson JD, O’Connor KA, Watkins LR, Maier SF (2004) The role of
Hanisch UK (2002) Microglia as a source and target of cytokines. Glia IL-1␤ in stress-induced sensitization of proinflammatory cytokine
40:140 –155. and corticosterone responses. Neuroscience 127:569 –577.
Harrison PJ (2002) The neuropathology of primary mood disorder. Kalivas PW, Nakamura M (1999) Neural systems for behavioral acti-
Brain 125:1428 –1449. vation and reward. Curr Opin Neurobiol 9:223–227.
Hashimoto K, Shimizu E, Iyo M (2004) Critical role of brain-derived Kamikawa H, Hori T, Nakane H, Aou S, Tashiro N (1998) IL-1beta
neurotrophic factor in mood disorders. Brain Res Rev 45:104 –114. increases norepinephrine level in rat frontal cortex: involvement of
Hauser P, Khosla J, Aurora H, Laurin J, Kling MA, Hill J, Gulati M, prostanoids, NO, and glutamate. Am J Physiol 275:R803–R810.
Thornton AJ, Schultz RL, Valentine AD, Meyers CA, Howell CD Keller MB (2002) Long-term treatment strategies in affective disorders.
(2002) A prospective study of the incidence and open-label treat- Psychopharmacol Bull 36:36 – 48.
ment of interferon-induced major depressive disorder in patients Kempermann G, Kronenberg G (2003) Depressed new neurons: adult
with Hepatitis C. Mol Psychiatry 7:942–947. hippocampal neurogenesis and a cellular plasticity hypothesis of
Hayley S, Brebner K, Lacosta S, Merali Z, Anisman H (1999) Sensi- major depression. Biol Psychiatry 54:499 –503.
tization to the effects of tumor necrosis factor-a: neuroendocrine, Kern W, Sieghart W (1994) Polyclonal antibodies directed against an
central monoamine and behavioral variations. J Neurosci 19: epitope specific for the alpha 4-subunit of GABAA receptors iden-
5654 –5665. tify a 67-kDa protein in rat brain membranes. J Neurochem
Hayley S, Kelly O, Anisman H (2003) Murine tumor necrosis factor ␣ 62:764 –769.
sensitizes plasma corticosoterone and manifestation of shock: Kling MA, Roy A, Doran AR, Calabrese JR, Rubinow DR, Whitfield HJ,
Modulation by histamine. J Neuroimmunol 131:60 – 69. May C, Post RM, Chrousos GP, Gold PW (1991) Cerebrospinal
S. Hayley et al. / Neuroscience 135 (2005) 659 – 678 675

fluid immunoreactive corticotropin-releasing hormone and adreno- Maes M, Meltzer HY (1995) The serotonin hypothesis of major de-
corticotropin secretion in Cushing’s disease and major depression: pression. In: Psychopharmacology: the third generation of
potential clinical implications. J Clin Endocrinol Metab 72: progress (Bloom FE, et al., eds), pp 993–944. New York: Raven
260 –271. Press.
Koch JM, Kell S, Hinze-Selch D, Aldenhoff JB (2002) Changes in Magarinos AM, McEwen BS, Flugge G, Fuchs E (1996) Chronic
CREB-phosphorylation during recovery from major depression. psychosocial stress causes apical dendritic atrophy of hippocam-
J Psychiatr Res 36:369 –375. pal CA3 pyramidal neurons in subordinate tree shrews. J Neurosci
Korpi ER, Kleinman JE, Wyatt RJ (1988) GABA concentrations in 16:3534 –3540.
forebrain areas of suicide victims. Biol Psychiatry 23:109 –114. Maher FO, Martin DS, Lynch MA (2004) Increased IL-1beta in cortex
Kraus MR, Schafer A, Faller H, Csef H, Scheurlen M (2002) Parox- of aged rats is accompanied by downregulatin of ERK and PI-3
etine for the treatment of interferon-alpha-induced depression in kinase. Neurobiol Aging 25(6):795– 806.
chronic hepatitis C. Aliment Pharmacol Ther 16:1091–1099. Maier SF, Watkins LR (1998) Cytokines for psychologists: Implications
Krystal JH, Sanacora G, Blumberg H, Anand A, Charney DS, Marek G, of bidirectional immune-to-brain communication for understanding
Epperson CN, Goddard A, Mason GF (2002) Glutamate and GABA
behavior, mood, and cognition. Psychol Rev 105:83–107.
systems as targets for novel antidepressant and mood-stabilizing
Malberg JE, Duman RS (2003) Cell proliferation in adult hippocampus
treatments. Mol Psychiatry 7:S71–S80.
is decreased by inescapable stress: reversal by fluoxetine treat-
Lai IC, Hong CJ, Tsai SJ (2003) Expression of cAMP response ele-
ment. Neuropsychopharmacology 28:1562–1571.
ment-binding protein in major depression before and after antide-
Malberg JE, Eisch AJ, Nestler EJ, Duman RS (2000) Chronic antide-
pressant treatment. Neuropsychobiology 48:182–185.
pressant treatment increases neurogenesis in adult rat hippocam-
Larson SJ, Romanoff RL, Dunn AJ, Glowa JR (2002) Effects of inter-
pus. J Neurosci 20:9104 –9110.
leukin-1beta on food-maintained behavior in the mouse. Brain
Behav Immun 16:398 – 410. Manji HK, Chen G (2002) PKC, MAP kinases and the bcl-2 family of
Leake A, Perry EK, Perry RH, Fairbairn AF, Ferrier IN (1990) Cortical proteins as long-term targets for mood stabilizers. Mol Psychiatry
concentrations of corticotropin-releasing hormone and its receptor 7:S46 –S56.
in Alzheimer type dementia and major depression. Biol Psychiatry Manji HK, Drevets WC, Charney DS (2001) The cellular neurobiology
28:603– 608. of depression. Nat Med 7:541–547.
Lemonde S, Turecki G, Bakish D, Du L, Hrdina PD, Bown CD, Se- Manji HK, Duman RS (2001) Impairments of neuroplasticity and cel-
queira A, Kushwaha N, Morris SJ, Basak A, Ou XM, Albert PR lular resilience in severe mood disorders: implications for the de-
(2003) Impaired repression at a 5-hydroxytryptamine 1A receptor velopment of novel therapeutics. Psychopharmacol Bull 35:5– 49.
gene polymorphism associated with major depression and suicide. Manji HK, Quiroz JA, Sporn J, Payne JL, Denicoff KA, Gray N, Zarate
J Neurosci 23:8788 – 8799. CA Jr., Charney DS (2003) Enhancing neuronal plasticity and
Linthorst ACE, Flachskamm C, Muller-Preuss P, Holsboer F, Reul cellular resilience to develop novel, improved therapeutics for dif-
JMHM (1995) Effect of bacterial endotoxin and interleukin-1b on ficult-to-treat depression. Biol Psychiatry 53:707–742.
hippocampal serotonergic neurotransmision, behavioral activity, Mann JJ, Huang YY, Underwood MD, Kassir SA, Oppenheim S, Kelly
and free corticosterone levels: An in vivo microdialysis study. TM, Dwork AJ, Arango V (2000) A serotonin transporter gene
J Neurosci 15:2920 –2934. promoter polymorphism (5-HTTLPR) and prefrontal cortical bind-
Liu D, Diorio J, Day JC, Francis DD, Meaney MJ (2000) Maternal care, ing in major depression and suicide. Arch Gen Psychiatry 57:
hippocampal synaptogenesis and cognitive development in rats. 729 –738.
Nat Neurosci 3:799 – 806. Marmigere F, Givalois L, Rage F, Arancibia S, Tapia-Arancibia L
Lowther S, De Paermentier F, Cheetham SC, Crompton MR, Katona (2003) Rapid induction of BDNF expression in the hippocampus
CL, Horton RW (1997) 5-HT1A receptor binding sites in post- during immobilization stress challenge in adult rats. Hippocampus
mortem brain samples from depressed suicides and controls. 13:646 – 655.
J Affect Disord 42:199 –207. Mattson MP, Maudsley S, Martin B (2004) BDNF and 5-HT: a dynamic
Lucassen PJ, Muller MB, Holsboer F, Bauer J, Holtrop A, Wouda J, duo in age-related neuronal plasticity and neurodegerative disor-
Hoogendijk WJ, De Kloet ER, Swaab DF (2001) Hippocampal ders. Trends Neurosci 27:589 –594.
apoptosis in major depression is a minor event and absent from McDonald WM, Richard IH, DeLong MR (2003) Prevalence, etiology,
subareas at risk for glucocorticoid overexposure. Am J Pathol and treatment of depression in Parkinson’s disease. Biol Psychia-
158:453– 468.
try 54:363–375.
Ma XC, Gottschall PE, Chen LT, Wiranowska M, Phelps CP (2002)
McEwen BS (2001) Plasticity of the hippocampus: adaptation to
Role and mechanisms of interleukin-1 in the modulation of neuro-
chronic stress and allostatic load. Ann N Y Acad Sci 933:265–277.
toxicity. Neuroimmunomodulation 10:199 –207.
Meaney MJ (2001) Maternal care, gene expression, and the transmis-
MacMaster FP, Kusumakar V (2004) Hippocampal volume in early
sion of individual differences in stress reactivity across genera-
onset depression. BMC Med 2:2
tions. Annu Rev Neurosci 24:1161–1192.
MacQueen GM, Campbell S, McEwen BS, Macdonald K, Amano S,
Menkes DB, MacDonald JA (2000) Interferons, serotonin and neuro-
Joffe RT, Nahmias C, Young LT (2003) Course of illness, hip-
pocampal function, and hippocampal volume in major depression. toxicity. Psychol Med 30:259 –268.
Proc Natl Acad Sci U S A 100:1387–1392. Merali Z, Brennan K, Brau P, Anisman H (2003) Dissociating anorexia
Maes M (1999) Major depression and activation of the inflammatory and anhedonia elicited by interleukin-1␤: antidepressant and gen-
response system. Adv Exp Med Biol 461:25– 46. der effects on responding for “free chow” and “earned” sucrose
Maes M, Bonaccorso S (2004) Lower activities of serum peptidases intake. Psychopharmacology 165:413– 418.
predict higher depressive and anxiety levels following interferon- Merali Z, Du L, Hrdina P, Palkovits M, Faludi G, Poulter MO, Anisman
alpha-based immunotherapy in patients with hepatitis C. Acta Psy- H (2004a) Dysregulation in the suicide brain: mRNA expression of
chiatr Scand 109:126 –131. corticotropin releasing hormone receptors and GABAA receptor
Maes M, Capuron L, Ravaud A, Gualde N, Bosmans E, Egyed B, et al. subunits in frontal cortical brain region. J Neurosci 24:1478 –1285.
(2001) Lowered serum dipeptidyl peptidase IV activity is associ- Merali Z, Khan S, Michaud DS, Shippy SA, Anisman H (2004b) Does
ated with depressive symptoms and cytokine production in cancer amygdaloid corticotropin-releasing hormone (CRH) mediate anxi-
patients receiving interleukin-2-based immunotherapy. Neuro- ety-like behaviors? Dissociation of anxiogenic effects and CRH
psychopharmacology 24:130 –140. release. Eur J Neurosci 20:229 –239.
676 S. Hayley et al. / Neuroscience 135 (2005) 659 – 678

Merali Z, Lacosta S, Anisman H (1997) Effects of interleukin-1b and electroconvulsive therapy. Corticotrophin-releasing factor,
mild stress on alterations of central monoamines: A regional mi- beta-endorphin and somatostatin. Br J Psychiatry 158:59 – 63.
crodialysis study. Brain Res 761:225–235. Nemeroff CB, Owens MJ, Bissette G, Andorn AC, Stanley M (1988)
Merali Z, McIntosh J, Kent P, Michaud D, Anisman H (1998) Aversive Reduced corticotropin releasing factor binding sites in the frontal
and appetitive events evoke the release of corticotropin-releasing cortex of suicide victims. Arch Gen Psychiatry 45:577–579.
hormone and bombesin-like peptides at the central nucleus of the Nemeroff CB, Widerlov E, Bissette G, Walleus H, Karlsson I, Eklund K,
amygdala. J Neurosci 18:4758 – 4766. Kilts CD, Loosen PT, Vale W (1984) Elevated concentrations of
Mercier G, Lennon AM, Renouf B, Dessouroux A, Ramauge M, Cour- CSF corticotropin-releasing factor-like immunoreactivity in de-
tin F, Pierre M (2004) MAP kinase activation by fluoxetine and its pressed patients. Science 226:1342–1344.
relation to gene expression in cultured rat astrocytes. J Mol Neu- Nguyen KT, Deak T, Owens SM, Kohno T, Fleshner M, Watkins LR,
rosci 24:207–216. Maier SF (1998) Exposure to acute stress induces brain interleu-
Merlo PE, Lorang M, Yeganeh M, Rodriguez DF, Raber J, Koob GF, kin-1␤ protein in the rat. J Neurosci 19:2799 –2805.
Weiss F (1995) Increase of extracellular corticotropin-releasing Nguyen PV, Woo NH (2003) Regulation of hippocampal synaptic
factor-like immunoreactivity levels in the amygdala of awake rats plasticity by cyclic AMP-dependent protein kinases. Prog Neuro-
during restraint stress and ethanol withdrawal as measured by biol 71:401– 437.
microdialysis. J Neurosci 15:5439 –5447. Obara Y, Nakahata N (2002) The signaling pathway of neurotrophic
Mertens S, Benke D, Mohler H (1993) GABAA receptor populations factor biosynthesis. Drug News Perspect 15:290 –298.
with novel subunit combinations and drug binding profiles identified Odagaki Y, Garcia-Sevilla JA, Huguelet P, La Harpe R, Koyama T,
in brain by alpha 5- and delta-subunit-specific immunopurification. Guimon J (2001) Cyclic AMP-mediated signaling components are
J Biol Chem 268:5965–5973. upregulated in the prefrontal cortex of depressed suicide victims.
Mervaala E, Fohr J, Kononen M, Valkonen-Korhonen M, Vainio P, Brain Res 898:224 –231.
Partanen K, Partanen J, Tiihonen J, Viinamaki H, Karjalainen AK, Pandey GN, Dwivedi Y, Rizavi HS, Ren X, Pandey SC, Pesold C,
Lehtonen J (2000) Quantitative MRI of the hippocampus and Roberts RC, Conley RR, Tamminga CA (2002) Higher expression
amygdala in severe depression. Psychol Med 30:117–125. of serotonin 5-HT(2A) receptors in the postmortem brains of teen-
Meyer JH, Kapur S, Houle S, DaSilva J, Owczarek B, Brown GM, age suicide victims. Am J Psychiatry 159:419 – 429.
Wilson AA, Kennedy SH (1999) Prefrontal cortex 5-HT2 receptors Pawate S, Shen Q, Fan F, Bhat NR (2004) Redox regulation of glial
in depression: an [18F]setoperone PET imaging study. Am J Psy- inflammatory response to lipopolysaccharide and interferon-
chiatry 156:1029 –1034. gamma. J Neurosci Res 77:540 –551.
Meyers CA, Valentine AD (1995) Neurological and psychiatric adverse Petty F (1995) GABA and mood disorders: a brief review and hypoth-
effects of immunological therapy. CNS Drugs 3:56 – 68. esis. J Affect Disord 34:275–281.
Minogue AM, Schmid AW, Fogarty MP, Moore AC, Campbell VA, Piao CS, Kim JB, Han PL, Lee JK (2003) Administration of the p38
Herron CE, Lynch MA (2003) Activation of the c-Jun N-terminal MAPK inhibitor SB203580 affords brain protection with a wide
kinase signaling cascade mediates the effect of amyloid-beta on therapeutic window against focal ischemic insult. J Neurosci Res
long term potentiation and cell death in hippocampus: a role for 73(4):537–544.
interleukin-1beta? J Biol Chem 278(30):27971–27980. Pierce RC, Pierce-Bancroft AF, Prasad BM (1999) Neurotrophin-3
Miyahara S, Komori T, Fujiwara R, Shizuya K, Yamamoto M, Ohmori contributes to the initiation of behavioral sensitization to cocaine by
M, Okazaki Y (2000) Effects of repeated stress on expression of activating the Ras/Mitogen-activated protein kinase signal trans-
interleukin-6 (IL-6) and IL-6 receptor mRNAs in rat hypothalamus duction cascade. J Neurosci 19:8685– 8695.
and midbrain. Life Sci 66: PL93–PL98. Pitts AF, Samuelson SD, Meller WH, Bissette G, Nemeroff CB, Kathol
Miyaoka H, Otsubo T, Kamijima K, Ishii M, Onuki M, Mitamura K RG (1995) Cerebrospinal fluid corticotropin-releasing hormone,
(1999) Depression from interferon therapy in patients with hepatitis vasopressin, and oxytocin concentrations in treated patients with
C. Am J Psychiatry 156:1120 major depression and controls. Biol Psychiatry 38:330 –335.
Moghaddam B, Bolinao ML, Stein-Behrens B, Sapolsky R (1994) Pocock JM, Liddle AC (2001) Microglial signalling cascades in neuro-
Glucocorticoids mediate the stress-induced extracellular accumu- degenerative disease. Prog Brain Res 132:555–565.
lation of glutamate. Brain Res 655:251–254. Pozzoli G, Bilezikjian LM, Perrin MH, Blount AL, Vale WW (1996)
Mullar N, Ackenheil M (1998) Psychoneuroimmunology and the cyto- Corticotropin-releasing factor (CRF) and glucocorticoids modulate
kine action in the CNS: implications for psychiatric disorders. Prog the expression of type 1 CRF receptor messenger ribonucleic acid
Neuropsychopharmacol Biol Psychiatry 22:1–33. in rat anterior pituitary cell cultures. Endocrinology 137:65–71.
Murphy PG, Borthwick LA, Altares M, Gauldie J, Kaplan D, Richardson Purselle DC, Nemeroff CB (2003) Serotonin transporter: a potential
PM (2000) Reciprocal actions of interleukin-6 and brain-derived substrate in the biology of suicide. Neuropsychopharmacology
neurotrophic factor on rat and mouse primary sensory neurons. 28:613– 619.
Eur J Neurosci 12:1891–1899. Raadsheer FC, Hoogendijk WJ, Stam FC, Tilders FJ, Swaab DF
Musselman DL, Lawson DH, Gumnick JF, Manatunga AK, Penna S, (1994) Increased numbers of corticotropin-releasing hormone ex-
Goodkin RS, Greiner K, Nemeroff CB, Miller AH (2001) Paroxetine pressing neurons in the hypothalamic paraventricular nucleus of
for the prevention of depression induced by high-dose interferon depressed patients. Neuroendocrinology 60:436 – 444.
alfa. N Engl J Med 344:961–966. Raadsheer FC, van Heerikhuize JJ, Lucassen PJ, Hoogendijk WJ,
Nagatsu T, Mogi M, Ichinose H, Togari A (2000) Changes in cytokines Tilders FJ, Swaab DF (1995) Corticotropin-releasing hormone
and neurotrophins in Parkinson’s disease. J Neural Transm Suppl mRNA levels in the paraventricular nucleus of patients with Alz-
60:277–290. heimer’s disease and depression. Am J Psychiatry 152:1372–
Nassberger L, Traskman-Bendz L (1993) Increased soluble interleu- 1376.
kin-2 receptor concentrations in suicide attempters. Acta Psychiatr Rage F, Givalois L, Marmigere F, Tapia-Arancibia L, Arancibia S
Scand 88:48 –52. (2002) Immobilization stress rapidly modulates BDNF mRNA ex-
Nemeroff CB (1996) The corticotropin-releasing factor (CRF) hypoth- pression in the hypothalamus of adult male rats. Neuroscience
esis of depression: New findings and new directions. Mol Psychi- 112:309 –318.
atry 1:336 –342. Rajkowska G (2000) Postmortem studies in mood disorders indicate
Nemeroff CB, Bissette G, Akil H, Fink M (1991) Neuropeptide concen- altered numbers of neurons and glial cells. Biol Psychiatry
trations in the cerebrospinal fluid of depressed patients treated with 48:766 –777.
S. Hayley et al. / Neuroscience 135 (2005) 659 – 678 677

Rasmusson AM, Shi L, Duman R (2002) Downregulation of BDNF Shaw KN, Commins S, O’Mara SM (2001) Lipopolysaccharide causes
mRNA in the hippocampal dentate gyrus after re-exposure to cues deficits in spatial learning in the watermaze but not in BDNF
previously associated with footshock. Neuropsychopharmacology expression in the rat dentate gyrus. Behav Brain Res 124:47–54.
27:133–142. Sheline YI, Gado MH, Kraemer HC (2003) Untreated depression and
Rattazzi M, Puato M, Faggin E, Bertipaglia B, Zambon A, Pauletto P hippocampal volume loss. Am J Psychiatry 160:1516 –1518.
(2003) C-reactive protein and interleukin-6 in vascular disease: Shen CP, Tsimberg Y, Salvadore C, Meller E (2004) Activation of Erk
culprits or passive bystanders? J Hypertens 21:1787–1803. and JNK MAPK pathways by acute swim stress in rat brain regions.
Reddy DS (2003) Is there a physiological role for the neurosteroid BMC Neurosci 20:5–36.
THDOC in stress-sensitive conditions? Trends Pharmacol Sci Sherif F, Marcusson J, Oreland L (1991) Brain gamma-aminobutyrate
24:103–106. transaminase and monoamine oxidase activities in suicide victims.
Reul JM, Holsboer F (2002) Corticotropin-releasing factor receptors 1 Eur Arch Psychiatry Clin Neurosci 241:139 –144.
and 2 in anxiety and depression. Curr Opin Pharmacol 2:23–33. Shiah IS, Yatham LN (1998) GABA function in mood disorders: an
Roach SK, Lee SB, Schorey JS (2005) Differential activation of the update and critical review. Life Sci 63:1289 –1303.
transcription factor cyclic AMP response element binding protein Shimizu E, Hashimoto K, Okamura N, Koike K, Komatsu N, Kumakiri
(CREB) in macrophages following infection with pathogenic and C, Nakazato M, Watanabe H, Shinoda N, Okada S, Iyo M (2003)
nonpathogenic mycobacteria and role for CREB in tumor necrosis Alterations of serum levels of brain-derived neurotrophic factor
factor alpha production. Infect Immun 73:514 –522. (BDNF) in depressed patients with or without antidepressants. Biol
Roth J, Harre EM, Rummel C, Gerstberger R, Hubschle T (2004) Psychiatry 54:70 –75.
Signaling the brain in systemic inflammation: role of sensory cir- Shintani F, Kanba S, Nakaki T, Nibuya M, Kinoshita N, Suzuki E, Yagi
cumventricular organs. Front Biosci 9:290 –300. G, Kato R, Asai M (1993) Interleukin-1␤ augments release of
Rothermundt M, Arolt V, Peters M, Gutbrodt H, Fenker J, Kersting A, norepinephrine, dopamine and serotonin in the rat anterior hypo-
et al. (2001) Inflammatory markers in major depression and mel- thalamus. J Neurosci 13:3574 –3581.
ancholia. J Affect Disord 63:93–102. Shintani F, Nakaki T, Kanba S, Sato K, Yagi G, Shiozawa M, Aiso S,
Rothwell NJ (1999) Annual review prize lecture cytokines: killers in the Kato R, Asai M (1995) Involvement of interleukin-1 in immobiliza-
brain? J Physiol 514:3–17. tion stress-induced increase in plasma adrenocorticotropic hor-
Roy A, Pickar D, Paul S, Doran A, Chrousos GP, Gold PW (1987) CSF mone and in release of hypothalamic monoamines in the rat.
J Neurosci 15:1961–1970.
corticotropin-releasing hormone in depressed patients and normal
Shirayama Y, Chen AC, Nakagawa S, Russell DS, Duman RS (2002)
control subjects. Am J Psychiatry 144:641– 645.
Brain-derived neurotrophic factor produces antidepressant effects
Roy-Byrne P, Wingerson DK, Radant A, Greenblatt DJ, Cowley DS
in behavioral models of depression. J Neurosci 22:3251–3261.
(1996) Reduced benzodiazepine sensitivity in patients with panic
Sibille E, Pavlides C, Benke D, Toth M (2000) Genetic inactivation of
disorder: comparison with patients with obsessive-compulsive dis-
the Serotonin(1A) receptor in mice results in downregulation of
order and normal subjects. Am J Psychiatry 153:1444 –1449.
major GABA(A) receptor alpha subunits, reduction of GABA(A)
Ruggiero DA, Underwood MD, Rice PM, Mann JJ, Arango V (1999)
receptor binding, and benzodiazepine-resistant anxiety. J Neurosci
Corticotropic-releasing hormone and serotonin interact in the hu-
20:2758 –2765.
man brainstem: behavioral implications. Neuroscience 91:1343–
Sibille E, Arango V, Galfalvy HC, Pavlidis P, Erraji-Benchekroun L,
1354.
Ellis SP, John Mann (2004) Gene expression profiling of depres-
Sapolsky RM (2000) The possibility of neurotoxicity in the hippocam-
sion and suicide in human prefrontal cortex. Neuropsychopharma-
pus in major depression: a primer on neuron death. Biol Psychiatry
cology 29:351–361.
48:755–765.
Silverstein FS, Barks JD, Hagan P, Liu XH, Ivacko J, Szaflarski J
Sapolsky RM, Krey LC, McEwen BS (1985) Prolonged glucocorticoid
(1997) Cytokines and perinatal brain injury. Neurochem Int 30:
exposure reduces hippocampal neuron number: implications for 375–383.
aging. J Neurosci 5:1222–1227. Sluzewska A (1999) Indicators of immune activation in depressed
Sapolsky RM, Uno H, Rebert CS, Finch CE (1990) Hippocampal patients. Adv Exp Med Biol 461:59 –74.
damage associated with prolonged glucocorticoid exposure in pri- Smith MA, Makino S, Altemus M, Michelson D, Hong SK, Kvetnansky
mates. J Neurosci 10:2897–2902. R, Post RM (1995) Stress and antidepressants differentially regu-
Saporito MS, Hudkins RL, Maroney AC (2002) Discovery of CEP- late neurotrophin 3 mRNA expression in the locus coeruleus. Proc
1347/KT-7515, an inhibitor of the JNK/SAPK pathway for the treat- Natl Acad Sci 92(19):8788 – 8792.
ment of neurodegenerative diseases. Prog Med Chem 40:23– 62. Smith RS (1991) The macrophage theory of depression. Med Hypoth-
Schatzberg AF, Schildkraut JJ (1995) Recent studies on norepineph- eses 35:298 –306.
rine systems on mood disorders. In: Psychopharmacology: the Sokoloff P, Guillin O, Diaz J, Carroll P, Griffon N (2002) Brain-derived
fourth generation of progress (Bloom FE, Kupfer DJ, eds), pp neurotrophic factor controls dopamine D3 receptor expression:
911–922. New York: Raven. implications for neurodevelopmental psychiatric disorders. Neuro-
Schlegel S, Steinert H, Bockisch A, Hahn K, Schloesser R, Benkert O tox Res 4:671– 678.
(1994) Decreased benzodiazepine receptor binding in panic disor- Song C, Merali Z, Anisman H (1999) Variations of nucleus accumbens
der measured by IOMAZENIL-SPECT. A preliminary report. Eur dopamine and serotonin following systemic interleukin-1, interleu-
Arch Psychiatry Clin Neurosci 244:49 –51. kin-2 or interleukin-6 treatment. Neuroscience 88:823– 836.
Schmidt ED, Binnekade R, Janszen AW, Tilders FJ (1996) Short Steiger JL, Russek SJ (2004) GABAA receptors: building the bridge
stressor induced long-lasting increases of vasopressin stores in between subunit mRNAs, their promoters, and cognate transcrip-
hypothalamic corticotropin-releasing hormone (CRH) neurons in tion factors. Pharmacol Ther 101:259 –281.
adult rats. J Neuroendocrinol 8:703–712. Stein-Behrens B, Mattson MP, Chang I, Yeh M, Sapolsky R (1994)
Schramm TM, Lawford BR, Macdonald GA, Cooksley WG (2000) Stress exacerbates neuron loss and cytoskeletal pathology in the
Sertraline treatment of interferon-alfa-induced depressive disorder. hippocampus. J Neurosci 14:5373–5380.
Med J Aust 173:359 –361. Stepanichev MY, Zdobnova IM, Yakovlev AA, Onufriev MV, Lazareva
Shanks N, Windle RJ, Perks PA, Harbuz MS, Jessop DS, Ingram CD, NA, Zarubenko II, Gulyaeva NV (2003) Effects of tumor necrosis
Lightman SL (2000) Early-life exposure to endotoxin alters hypotha- factor-alpha central administration on hippocampal damage in rat
lamic-pituitary-adrenal function and predisposition to inflammation. induced by amyloid beta-peptide (25–35). J Neurosci Res
Proc Natl Acad Sci U S A 97:645–650. 71:110 –120.
678 S. Hayley et al. / Neuroscience 135 (2005) 659 – 678

Stevenson CW, Sullivan RM, Gratton A (2003) Effects of basolateral tein overexpression in the basolateral amygdala on behavioral
amygdala dopamine depletion on the nucleus accumbens and models of depression and anxiety. Biol Psychiatry 56:151–160.
medial prefrontal cortical dopamine responses to stress. Neuro- Wang CX, Shuaib A (2002) Involvement of inflammatory cytokines in
science 116:285–293. central nervous system injury. Prog Neurobiol 67:161–172.
Stockmeier CA (2003) Involvement of serotonin in depression: evi- Wang Q, Walsh DM, Rowan MJ, Selkoe DJ, Anwyl R (2004) Block of
dence from postmortem and imaging studies of serotonin recep- long-term potentiation by naturally secreted and synthetic amyloid
tors and the serotonin transporter. J Psychiat Res 37:357–373. beta-peptide in hippocampal slices is mediated via activation of the
Stockmeier CA, Dilley GE, Shapiro LA, Overholser JC, Thompson PA, kinases c-Jun N-terminal kinase, cyclin-dependent kinase 5, and
Meltzer HY (1997) Serotonin receptors in suicide victims with major p38 mitogen-activated protein kinase as well as metabotropic glu-
depression. Neuropsychopharmacology 16:162–173. tamate receptor type 5. J Neurosci 24(13):3370 –3378.
Stoll G, Jander S, Schroeter M (2000) Cytokines in CNS disorders: Wichers M, Maes M (2002) The psychoneuroimmuno-pathophysiology
neurotoxicity versus neuroprotection. J Neural Transm 59:81– 89. of cytokine-induced depression in humans. Int J Neuropsychop-
Stout SC, Owens MJ, Lindsey KP, Knight DL, Nemeroff CB (2001) harmacol 5:375–388.
Effects of sodium valproate on corticotropin-releasing factor sys- Wichers MC, Maes M (2004) The role of indoleamine 2,3-dioxygenase
tems in rat brain. Neuropsychopharmacology 24:624 – 631. (IDO) in the pathophysiology of interferon-alpha-induced depres-
Sundman-Eriksson I, Allard P (2002) [(3)H]Tiagabine binding to GABA sion. J Psychiatry Neurosci 29:11–17.
transporter-1 (GAT-1) in suicidal depression. J Affect Disord 71: Wise RA (2004) Dopamine, learning and motivation. Nat Rev Neurosci
29 –33. 5:483– 494.
Sundman I, Allard P, Eriksson A, Marcusson J (1997) GABA uptake Wong ML, Kling MA, Munson PJ, Listwak S, Licinio J, Prolo P, Karp B,
sites in frontal cortex from suicide victims and in aging. Neuropsy- McCutcheon IE, Geracioti TD Jr, DeBellis MD, Rice KC, Goldstein
chobiology 35:11–15. DS, Veldhuis JD, Chrousos GP, Oldfield EH, McCann SM, Gold
Tan H, Zhong P, Yan Z (2004) Corticotropin-releasing factor and acute PW (2000) Pronounced and sustained central hypernoradrenergic
stress prolongs serotonergic regulation of GABA transmission in function in major depression with melancholic features: relation to
hypercortisolism and corticotropin-releasing hormone. Proc Natl
prefrontal cortical pyramidal neurons. J Neurosci 24:5000 –5008.
Acad Sci U S A 97:325–330.
Tanji J, Hoshi E (2001) Behavioral planning in the prefrontal cortex.
Wrynn AS, Sebens JB, Koch T, Leonard BE, Korf J (2000) Prolonged
Curr Opin Neurobiol 11:164 –170.
c-Jun expression in the basolateral amygdala following
Tilders FJH, Schmidt ED, De Goeij DCE (1993) Phenotypic plasticity
bulbectomy: possible implications for antidepressant activity and
of CRF neurons during stress. Ann N Y Acad Sci 697:39 –52.
time of onset. Brain Res Mol Brain Res 76(1):7–17.
Tilders FJH, Schmidt ED (1999) Cross-sensitization between immune
Yamada S, Yamamoto M, Ozawa H, Riederer P, Saito T (2003)
and non-immune stressors. A role in the etiology of depression?
Reduced phosphorylation of cyclic AMP-responsive element bind-
Adv Exp Med Biol 461:179 –197.
ing protein in the postmortem orbitofrontal cortex of patients with
Tunnicliff G, Malatynska E (2003) Central GABAergic systems and
major depressive disorder. J Neural Transm 110(6):671– 680.
depressive illness. Neurochem Res 28:965–976.
Yan Z, Flores-Hernandez J, Surmeier DJ (2001) Coordinated expres-
Uno H, Eisele S, Sakai A, Shelton S, Baker E, DeJesus O, Holden J
sion of muscarinic receptor messenger RNAs in striatal medium
(1994) Neurotoxicity of glucocorticoids in the primate brain. Horm
spiny neurons. Neuroscience 103:1017–1024.
Behav 28:336 –348.
Yehuda R (2002) Post-traumatic stress disorder. N Engl J Med
Vakili K, Pillay SS, Lafer B, Fava M, Renshaw PF, Bonello-Cintron CM, 346(2):108 –114.
Yurgelun-Todd DA (2000) Hippocampal volume in primary unipolar Ying SW, Futter M, Rosenblum K, Webber MJ, Hunt SP, Bliss TV,
major depression: a magnetic resonance imaging study. Biol Psy- Bramham CR (2002) Brain-derived neurotrophic factor induces
chiatry 47:1087–1090. long-term potentiation in intact adult hippocampus: requirement for
Valentine AD, Meyers CA, Talpaz M (1995) Treatment of neurotoxic ERK activation coupled to CREB and upregulation of Arc synthe-
side effects of interferon-alpha with naltrexone. Cancer Invest sis. J Neurosci 22:1532–1540.
13:561–566. Yirmiya R, Weidenfeld J, Pollak Y, Morag M, Morag A, Avitsur R, Barak
Varfolomeev EE, Ashkenazi A (2004) Tumor necrosis factor: an apo- O, Reichenberg A, Cohen E, Shavit Y, Ovadia H (1999) Cytokines,
ptosis JuNKie? Cell 116:491– 497. “depression due to a general medical condition,” and antidepres-
Vezzani A, Moneta D, Richichi C, Aliprandi M, Burrows SJ, Ravizza T, sant drugs. Adv Exp Med Biol 461:283–316.
Perego C, De Simoni MG (2002) Functional role of inflammatory Zanardi R, Artigas F, Moresco R, Colombo C, Messa C, Gobbo C,
cytokines and antiinflammatory molecules in seizures and epilep- Smeraldi E, Fazio F (2001) Increased 5-hydroxytryptamine-2 re-
togenesis. Epilepsia 43:30 –35. ceptor binding in the frontal cortex of depressed patients respond-
Vythilingam M, Vermetten E, Anderson GM, Luckenbaugh D, Ander- ing to paroxetine treatment: a positron emission tomography scan
son ER, Snow J, Staib LH, Charney DS, Bremner JD (2004) study. J Clin Psychopharmacol 21:53–58.
Hippocampal volume, memory, and cortisol status in major depres- Zhang D, Kishihara K, Wang B, Mizobe K, Kubo C, Nomoto K (1998)
sive disorder: effects of treatment. Biol Psychiatry 56:101– Restraint stress-induced immunosuppression by inhibiting leuko-
112. cyte migration and Th1 cytokine expression during the intraperito-
Walker DL, Davis M (1997) Double dissociation between the involve- neal infection of Listeria monocytogenes. J Neuroimmunol 92:
ment of the bed nucleus of the stria terminalis and the central 139 –151.
nucleus of the amygdala in startle increases produced by condi- Ziemssen T, Kumpfel T, Klinkert WE, Neuhaus O, Hohlfeld R (2002)
tioned versus unconditioned fear. J Neurosci 17:9375–9383. Glatiramer acetate-specific T-helper 1- and 2-type cell lines pro-
Wallace TL, Stellitano KE, Neve RL, Duman RS (2004) Effects of duce BDNF: implications for multiple sclerosis therapy. Brain-de-
cyclic adenosine monophosphate response element binding pro- rived neurotrophic factor. Brain 125:2381–2391.

(Accepted 22 March 2005)


(Available online 8 September 2005)

Você também pode gostar