Вы находитесь на странице: 1из 10

BRAIN,

BEHAVIOR,
and IMMUNITY
Brain, Behavior, and Immunity 21 (2007) 374–383
www.elsevier.com/locate/ybrbi

Named Series: Twenty Years of Brain, Behavior, and Immunity

Depressive disorders and immunity: 20 years of progress and discovery


a,* b
Michael R. Irwin , Andrew H. Miller
a
Cousins Center for Psychoneuroimmunology, University of California, Los Angeles, Semel Institute for Neuroscience, 300 Medical Plaza, Suite 3-109,
Los Angeles, CA, USA
b
Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA

Received 28 October 2006; received in revised form 10 January 2007; accepted 12 January 2007
Available online 13 March 2007

Abstract

Since the inception of Brain, Behavior and Immunity twenty years ago, many exciting developments have occurred regarding the rela-
tionship between depression and the immune system. These developments have increasingly put the field of psychoneuroimmunology
into a clinical context with important translational implications. Initial studies focused on the impact of depression on relatively narrowly
defined immunologic endpoints, which ultimately found their relevance in studies examining the effect of depression on immunologically-
based diseases including infectious illnesses, autoimmune disorders, and cancer as well as more recently cardiovascular disease. Mech-
anistic studies have also greatly contributed to an understanding of those facets of depression, which might mediate these effects. More
recently, the reciprocal influences of the immune system on the brain and behavior including depression have taken center stage. Increas-
ing data now indicate that activated inflammatory processes can influence multiple aspects of CNS function including neurotransmitter
metabolism, neuroendocrine function, and information processing leading to behavioral changes in humans that bespeak depression.
These latter developments have intrigued scientists investigating the pathophysiology of depression and warrant consideration as some
of the most exciting new developments in psychiatry in the past 20 years. What the future holds is a world of promise as multiple trans-
lational targets derived from the cytokine model of depression work their way into the clinical arena as drug targets for further devel-
opment. Moreover, the work has served to instantiate brain-immune interactions as an essential component in psychiatric and medical
co-morbidities and their impact on health and illness.
Ó 2007 Elsevier Inc. All rights reserved.

1. Introduction ably occur in patients with major depressive disorder (Zor-


rilla et al., 2001). During the first ten years of Brain
Depression has a huge impact on individuals and soci- Behavior, and Immunity (BBI), a number of immune find-
ety. With a lifetime prevalence of over 15%, depression will ings were identified in major depression. Further studies
be the second leading illness in the world by 2020 as pro- published during this decade and beyond began to consider
jected by the World Health Organization. In addition to the behavioral correlates and biological mechanisms that
the emotional consequences of depression, this disorder is might be involved; these data are conceptually grouped
increasingly implicated in a wide range of medical condi- together in the first decade. Over the second ten years of
tions. Moreover, a growing body of evidence indicates that BBI, evidence emerged that depression is also associated
depression, even minor depression, has notable immuno- with activation of the innate inflammatory immune
logical consequences. As recently described in a compre- response including alterations in the ability of immune cells
hensive meta-analysis of over 180 studies with more than to express proinflammatory cytokines. Recent interest has
40 immune measures, many immunological changes reli- focused on the hypothesis that these cytokine abnormali-
ties may have reciprocal influences on the central nervous
system and contribute in part to the pathophysiology of
*
Corresponding author. Fax: +1 310 794 9247. the disorder. As BBI moves into its third decade, future
E-mail address: mirwin1@ucla.edu (M.R. Irwin). research directions are discussed with an emphasis on the

0889-1591/$ - see front matter Ó 2007 Elsevier Inc. All rights reserved.
doi:10.1016/j.bbi.2007.01.010
M.R. Irwin, A.H. Miller / Brain, Behavior, and Immunity 21 (2007) 374–383 375

Fig. 1. Brain–immune interactions in depression. The experience of depression on the immune system is moderated by age, sex, socioeconomic status, life
stress, physical activity, and sleep. In turn, the central release of corticotropin releasing hormone in depressed persons activates the hypothalamic pituitary
adrenal axis and the sympathetic nervous system. For these efferent pathways, multiple aspects of the immune system are altered with evidence of immune
suppression (e.g., decreases in lymphocyte responses), as well as inflammation. In turn, such immune changes have implications for infectious disease risk
and the occurrence of inflammatory disorders.

emerging evidence that immune alterations in depression and function of immune cells might be a mechanism to
have clinical implications not only as targets for the treat- account for illness in depressed persons.
ment of depression, but also as relevant risk factors for the Among the first immunological changes identified in
development and progression of infectious diseases and depressed persons were increases in the total number of
other immunologically based disorders (Fig. 1). white blood cells and in the numbers and percentages of
neutrophils and lymphocytes (Zorrilla et al., 2001). Enu-
meration of lymphocyte subsets also revealed that depres-
2. In the beginning . . . 1987 sion was associated with decreases in the number and
percentage of lymphocytes (B cells, T cells, T helper cells
2.1. Immune findings in major depression and T suppressor/cytotoxic cells), as well as a decrease in
the circulating number of cells that express the NK cell
In an effort to understand whether brain and behavior phenotype (Zorrilla et al., 2001). However, in one of the
had a role in the regulation of immunity in humans, major largest study samples of depressed subjects, no difference
depression served as an excellent clinical model to learn in the number of peripheral blood lymphocytes or T-lym-
more about brain–behavior–immune interactions as phocyte subsets was found between depressed patients
depressed patients exhibit prominent abnormalities of and controls (Schleifer et al., 1989). Moreover, meta-ana-
behavior (e.g., depressed mood, impaired sleep), along with lytic findings also questioned whether there are consistent
dysregulation of the neuroendocrine- and sympathetic ner- changes in the number of circulating B, T, or NK cells in
vous systems (SNS), which were identified as key efferent depression (Zorrilla et al., 2001). Given the heterogeneity
pathways in the regulation of immunity by the brain. In in depressed patient populations, these data suggested that
addition, given the significant health consequences of moderating clinical and biological factors may account for
depression, it was hypothesized that changes in the number the lack of consistency in results (see below).
376 M.R. Irwin, A.H. Miller / Brain, Behavior, and Immunity 21 (2007) 374–383

Early studies also examined functional immune 2.3. Effects of clinical treatment
measures, with a majority assaying non-specific mitogen-
induced lymphocyte proliferation. As compared to Only a limited number of studies have investigated the
controls, depressed patients were reported to show clinical course of depression and changes of cellular
decreases in lymphocyte proliferative responses (Zorrilla immunity in relation to antidepressant medication treat-
et al., 2001). Some studies, however, failed to replicate ment and symptom resolution. In one longitudinal
these observations (Schleifer et al., 1989), which might have case-control study, depressed patients showed an increase
been accounted for by a failure to control for day-to-day in NK activity during a 6-month course of tricyclic anti-
assay variability in the comparison of depressed patients depressant medication treatment and symptom resolu-
vs. matched controls (Schleifer et al., 1993). More than a tion, with improvements correlated with declines of
dozen studies have now been conducted on lymphocyte symptom severity and not treatment status at the time
proliferation in depression, and there is a reliable associa- of follow-up (Irwin et al., 1992). In another longitudinal
tion between depression and decreased proliferative follow-up study of young adults with unipolar depression
responses to the three non-specific mitogens including involving 6 weeks of treatment with nortriptyline and
phytohaemaglutinin (PHA), concanavalin-A (Con A), alprazolam, clinical improvements in the severity of
and pokeweed (PWM) (Zorrilla et al., 2001). depressive symptoms was associated with decreased num-
In the inaugural issue of BBI, it was reported that sever- bers of circulating lymphocytes and decreased responses
ity of depressive symptoms in persons undergoing severe to PHA and Con A but not PWM (Schleifer et al.,
life stress was associated with declines in another func- 1999). In addition, decreases in T cells, CD4, and
tional measure of the immune system, natural killer cell CD29 were found although there were no changes in B
(NK) activity (Irwin et al., 1987a). During the next ten cell numbers or CD8 cells. None of these changes were
years, a number of studies extended this observation to related to nortriptyline blood levels. In addition,
major depression; a reduction of NK activity is now viewed in vivo and in vitro treatment with fluoxetine, a selective
as one of the most reliable immune alterations found in this serotonin reuptake inhibitor, resulted in enhanced NK
disorder (Zorrilla et al., 2001). activity along with changes in depressive symptoms
(Frank et al., 1999), consistent with the finding of
2.2. Clinical moderating factors Ravidran et al. in which a number of different antide-
pressants were used including nafazodone, paroxetine,
It has been recognized that several clinical variables may sertraline, venlafaxine (Ravindran et al., 1995). Together,
influence immune measures and moderate the association these data suggest that a resolution of depressive symp-
between depression and changes in enumerative and func- toms results in improvements in lymphocyte responses
tional measures of immunity, including age, sex, body and NK activity, which are not simply due to effects of
mass, life stress, smoking and other psychiatric co-morbid- antidepressant medications on immune cell function.
ity (Stein et al., 1991). For example, older adults show
declines in cellular immunity, and it appears that the pres- 2.4. Biological mediators
ence of depression exacerbates age-related immune altera-
tions (Schleifer et al., 1989; Andreoli et al., 1993). 2.4.1. Corticotropin releasing hormone
Second, as compared to depressed women, declines of T Depressed patients show elevated levels of corticotro-
cell and NK cell responses are more prominent in pin releasing hormone (CRH) in the central nervous sys-
depressed men (Evans et al., 1992). Third, in healthy adult tem as measured by radioimmunoassay of cerebrospinal
depressed patients, adiposity and larger body mass par- fluid (Nemeroff et al., 1984), and this key neuropeptide
tially mediate the increase of inflammatory markers (see is involved in integrating neural, neuroendocrine, as well
below) (Miller et al., 2002b). Fourth, life stress can also as immune responses to stress. In the first issue of BBI,
alter functional immune measures, and depressed patients animal models were used to show that central doses of
who evidence greater life stress show greater declines of CRH induced marked declines of NK activity (Irwin
NK activity than depressed patients who are not stressed et al., 1987b), and that this response was mediated by cen-
(Irwin et al., 1990c). Fifth, in regards to smoking, a large tral CRH receptor mechanisms which were independent
study of 245 depressed and comparison controls stratified of the release of adrenocorticotropic hormone (ACTH)
by smoking status found that depression and smoking sta- and cortisol (Irwin et al., 1990b,d). Further studies
tus interact to produce greater declines of NK activity than showed that central doses of CRH induced decreases of
changes found in depressed or smoking groups alone (Jung cellular and humoral immune responses, with sympathetic
and Irwin, 1999). Finally, specific diagnostic comorbidity effector mechanisms mediating the action of central CRH
such as anxiety disorder and alcohol dependence (Irwin on the immune system (Strausbaugh and Irwin, 1992;
et al., 1990a; Schleifer et al., 2006) in combination with Friedman and Irwin, 2001). These data implicated central
depression induce greater declines of NK activity than CRH as being a key neuropeptide that might coordinate
changes in depressed patients without such co-morbid and induce immune suppression characterized in
histories. depressed patients.
M.R. Irwin, A.H. Miller / Brain, Behavior, and Immunity 21 (2007) 374–383 377

2.4.2. Sympathetic effector mechanisms peripheral blood mononuclear cells in patients with major
Given emerging data that sympathetic effector mecha- depression (Maes et al., 1993).
nisms have potent suppressive effects on natural and cellu-
lar immune responses in animal models, attention began to 2.5. Behavioral mechanisms
focus on the role of sympathetic neurotransmitters in con-
tributing to the alterations of immune function in depres- In addition to the biological mediators of immune
sion. At rest and in response to acute physical and/or changes in depression, several behavioral factors associated
psychological challenge, depressed patients show elevated with depression appear to contribute to immune dysfunc-
levels of circulating catecholamines and neuropeptide Y tion. Indeed, as reviewed by Cohen and Miller (Miller
(Irwin et al., 1991), and this activation uniquely predicted et al., 1999), examination of behavioral factors is needed
declines of NK cell responses. Sympathetic nerve terminals in clinical psychoneuroimmunology.
are juxtaposed with immune cells in organs where immune
system cells develop and respond to pathogens (Sanders 2.5.1. Physical activity and exercise as moderators
and Straub, 2002). When sympathetic release of norepi- Physical activity, or a lack thereof, can have negative con-
nephrine and neuropeptide Y occurs, sympathetic neuro- sequences on the immune system. Conversely, exercise has
transmitter receptor binding serves as a signal in a been shown to have potent salutary effects on immune mea-
connection between the brain and the immune system sures such as NK activity (Nieman et al., 1990). Hence, a few
and leads to suppression of NK and cellular immune studies have explored whether changes in physical activity
responses (Sanders and Straub, 2002), which is a more might moderate alterations in immunity among depressed
direct and specific influence than that provided by circulat- patients. In the meta-analysis of Herbert and Cohen, melan-
ing mediators. cholic depression correlated with greater impairments of cel-
lular immunity, which may be due, at least in part, to an
increased predominance of neurovegetative symptoms (Her-
2.4.3. Neuroendocrine axis bert and Cohen, 1993). Irwin found that severity of psycho-
A hallmark of major depression is dysregulation of the motor retardation uniquely predicted declines of NK
hypothalamic pituitary adrenal axis (HPA) and the hyper- activity in depression (Cover and Irwin, 1994).
secretion of cortisol. Cortisol exerts diverse effects on a
wide variety of physiological systems, and also coordinates 2.5.2. Insomnia and disordered sleep
the action of various cells involved in an immune response A number of studies have also shown that sleep has a
by altering cell distribution and the production of cyto- potent role in the behavioral regulation of immune func-
kines or immune messengers. Similar to sympathetic acti- tion (Irwin et al., 1996). Given that insomnia is one of
vation, cortisol can suppress the cellular immune the most common complaints of depressed subjects, inves-
response critical in defending the body against viral infec- tigators explored whether sleep disturbances were associ-
tions, possibly by increasing serum soluble CD8 or sup- ated with immune alterations in depression. Decreases of
pressor/cytotoxic antigen concentrations (Maes et al., NK activity were found to correlate with subjective insom-
1996). Cortisol can also prompt some immune cells to nia, but not with other depressive symptoms including
move out from circulating blood into lymphoid organs or somatization, weight loss, cognitive disturbance, or diurnal
peripheral tissues such as the skin. variation in depressed patients (Irwin, 2002). Likewise, in
Despite these data, a relationship between HPA axis studies that evaluated sleep using polysomnography,
activation and cellular immune alterations in depression decreases of total sleep as well as sleep efficiency were
has not been convincingly demonstrated. For example, in shown to correlate with declines in natural and cellular
depressed patients, decreased lymphocyte responses to mit- immune function among depressed patients. Further stud-
ogens are neither associated with dexamethasone nonsup- ies involving subjects with primary insomnia (e.g., no
pression nor with increased excretion rates of urinary free depression, other psychiatric disorder, and medical disor-
cortisol (Kronfol and House, 1985; Kronfol et al., 1986), der) have found that prolonged sleep latency and fragmen-
although HPA-axis non-suppressors exhibited a relative tation of sleep are associated with nocturnal elevations of
resistance to the enhancing (e.g. neutrophils) or depressant sympathetic catecholamines and declines in daytime levels
(e.g. lymphocytes, CD4 T cells) effects of dexametha- of NK cell responses (Irwin et al., 2003).
sone(Maes et al., 1994). Dexamethasone nonsuppression
refers to the diminished feedback inhibition of the HPA 3. The second decade . . . 1997
axis, and is typically associated with HPA hyperactivity.
Of note, however, as indicated below, dexamethasone 3.1. Viral specific and disease-relevant in vivo immune
non-suppression has been associated with evidence of measures
increased activation of innate immune responses. For
example, a significant correlation has been reported In a provocative review of the immune findings in
between post dexamethasone cortisol levels and increased depression, the significance and clinical meaning of non-
mitogen-induced interleukin-1 b (IL-1) responses of specific immune alterations (i.e., changes in immune cell
378 M.R. Irwin, A.H. Miller / Brain, Behavior, and Immunity 21 (2007) 374–383

distribution, NK activity and lymphocyte proliferation) and T helper 1 vs. T helper 2 (Th1/Th2) cytokines that were
was challenged (Stein et al., 1991). Thus, investigators were involved in initiating and coordinating immune responses
spurred to evaluate more salient immune responses that to infectious challenge (Moynihan et al., 1990). Consistent
might be relevant to risk of disease. With in vivo challenge with these basic findings, lipopolysaccharide-stimulated
by means of delayed type hypersensitivity (DTH) production of IL-1 and interleukin-6 (IL-6) was increased
responses, it was reported that depressed patients were less in depressed patients, and the relative balance of Th1/
responsive to a panel of antigenic challenges. Moreover, Th2 cytokines was found to be shifted toward an increase
memory T cell function in response to a specific viral path- in the capacity of lymphocytes to produce the Th1 cytokine
ogen was lower in depressed patients as compared to con- interferon in depression (Seidel et al., 1995). Although no
trols (Irwin et al., 1998). In other studies, psychological difference in stimulated production of IL-2 was found (Sei-
stress was reported to induce a marked decline in specific del et al., 1995; Irwin et al., 2003), it has been suggested
immune responses to immunization against viral infections that the capacity of lymphocytes to produce IL-2 may dif-
(Vedhara et al., 1999), although extension of this work to fer between melancholic and non-melancholic depression
major depression has yet to be performed. (Schlatter et al., 2004). In addition, peripheral blood mono-
To complement these more disease relevant immuno- nuclear cells of non-melancholic depressed patients showed
logic endpoints, studies also began to focus on the impact a greater stimulated capacity to produce IL-1 and IL-1
of depression on immunologically based diseases, including receptor antagonist as compared to responses from con-
infectious diseases. For example, HIV infection shows a trols and melancholic depressed patients (Kaestner et al.,
highly variable course, and depression, bereavement, and 2005), and serum levels of IL-1 were reported to be ele-
maladaptive coping responses to stress (including the stress vated in patients with late-life depression (Thomas et al.,
of HIV infection itself) have all been shown to predict the 2005).
rate of immune system decay in HIV patients (Cruess et al.,
2005). Indeed, major depression in women with HIV infec- 3.3. Inflammation and circulating levels of inflammatory
tion has been associated with lower NK activity, as well as markers
increases in the numbers of activated CD8 lymphocytes
and viral load, which suggested that declines of killer lym- In patients with inflammatory disorders such as rheu-
phocytes in association with depression may increase risk matoid arthritis (Zautra et al., 2004) and cardiovascular
of HIV disease progression in women (Cruess et al., disease (Lesperance et al., 2004), depression has been
2005). Immune system decline and HIV replication are par- found to predict increased morbidity and mortality, a
ticularly rapid in patients living under chronic stress (e.g., finding potentially related to the contribution of depres-
gay men who conceal their homosexuality by living ‘‘in sion to immune activation and inflammation. Indeed,
the closet’’) and in patients with high levels of SNS activity depression has been associated with increases in circulat-
(e.g., socially inhibited introverts). Tissue culture studies ing levels of the pro-inflammatory cytokine, IL-6 in adults
have shown that SNS neurotransmitters and glucocorti- with major depression (Zorrilla et al., 2001), in depressed
coids can accelerate HIV replication by rendering T lym- elderly populations, and in those depressed persons with
phocytes more vulnerable to infection and by suppressing chronic medical disorders such as rheumatoid arthritis
production of the anti-viral cytokines that help cells limit (Zautra et al., 2004), cancer (Musselman et al., 2001),
viral replication (Cole et al., 2001). Together, these data and cardiovascular disease (Lesperance et al., 2004).
suggest that life stress may play a role in the effects of Recent data further show that depressed patients show
depression on the immune system. an exaggerated activation of the inflammatory response
In addition to viral infections, experimental studies con- following acute psychological stress, with greater increases
ducted in animal models have implicated decreases in NK of IL-6 as well as activation of nuclear factor (NF)-jB, a
cell function in facilitating the metastatic spread of NK cell transcription factor that signals the inflammatory cascade
sensitive tumors (Page et al., 1994). However, translation (Pace et al., 2006). Moreover, depressed patients with
of these findings to the clinic has been challenging, with more severe sleep disturbance may be at greater risk for
limited support for a link between depression and cancer elevated levels of IL-6 and other proinflammatory mark-
(Antoni et al., 2006). Nevertheless, in patients with meta- ers, because sleep loss has been shown to induce acute
static malignant melanoma, group psychotherapy led to increases in cellular and genomic markers of inflammation
improvements in depressed mood, increases in NK cyto- (Irwin et al., 2006).
toxicity, and increased survival time, controlling for initial Increases in circulating levels of other proinflammatory
staging and medical care during the follow-up period (Faw- cytokines such as tumor necrosis factor a (TNF) and IL-1
zy et al., 2003). have also been reported in depressed patients (Anisman
et al., 1999) including late-life depressive disorder.
3.2. Stimulated cytokine production Increases of plasma levels of IL-12 in a large cohort of
depressed patients have also been found. IL-12 is a hetero-
In animal models, foot-shock stress was found to alter dimeric cytokine that is produced primarily by monocytes
the expression of pro-inflammatory, anti-inflammatory, and macrophages and plays a central role in the early
M.R. Irwin, A.H. Miller / Brain, Behavior, and Immunity 21 (2007) 374–383 379

phases of inflammation. Although early reports suggested 3.4.1. Depression in the medically ill
increases in haptoglobin as well as some other acute phase Major depression is frequent in the medically ill with up
proteins in depressed patients (Zorrilla et al., 2001), these to 50% of patients with medical illnesses exhibiting depres-
data were primarily limited to reports from one laboratory sive symptoms, depending on the type and severity of dis-
and recent efforts have failed to identify abnormal increases ease. New developments in the biology of mood disorders
in acute phase proteins (Pike and Irwin, 2006) consistent have raised the possibility that proinflammatory cytokines,
with the preliminary conclusions from meta-analyses (Zor- released as a function of disease related inflammatory pro-
rilla et al., 2001). Nevertheless, increases in c-reactive pro- cesses, participate in the pathophysiology of depression.
tein have been found in association with depression with Indeed, given evidence that cytokines induce a syndrome
elevations in healthy depressed adults (Miller et al., referred to as ‘‘sickness behavior’’ that has many overlap-
2002b), as well as in those depressed patients with acute ping features with major depression, cytokine-induced sick-
coronary syndrome (Lesperance et al., 2004). In turn, sys- ness behavior may account in part for the high rate of
temic immune activation is thought to lead to endothelial major depression found in the medically ill. Relevant to
activation in depression with increases in the expression its mediation by proinflammatory cytokines, sickness
of soluble intercellular adhesion molecule (sICAM) (Les- behavior can be reliably reproduced by administration of
perance et al., 2004). each of the proinflammatory cytokines in isolation or by
Depression appears to yield increases in inflammatory administering agents (e.g. endotoxin or lipopolysaccharide
markers in some patients, and decreases of NK responses [LPS]) that induce the proinflammatory cytokine cascade
in other patients (Pike and Irwin, 2006). It is not known (TNF-a to IL-1 to IL-6) (Yirmiya et al., 1999). Relevant
what mechanisms might account for the dissociation to the role of cytokines in behavioral pathology in medi-
between inflammation and innate immune measures in cally ill patients, depressed patients with cancer were found
depression, although genetic and metabolic variation in to exhibit significantly higher plasma IL-6 concentrations
the expression of proinflammatory cytokines may play compared to non-depressed cancer patients and healthy
a role. For example, stress-induced increases of plasma controls (Musselman et al., 2001). In addition, elevated
C-reactive protein is reported to occur only in stressed plasma concentrations of cytokines have been found in
persons who have the A allele of tumor necrosis factor association with specific symptoms of depression, including
a -308 G/A polymorphism. Likewise, polymorphism of elevated production and circulating levels inflammatory
the 174 bp upstream of the transcription initiation site markers in cancer patients with significant fatigue (Colla-
of the IL6 gene, the –174G/C allele, correlates with do-Hidalgo et al., 2006) and elevated IL-6 concentrations
plasma IL6 levels, although the relationship of this poly- in cancer patients with impaired executive function.
morphism with major depression is not known. Finally,
one third of total IL-6 in the circulation is estimated 3.4.2. Cytokine therapies
to originate from adipose tissue, and even in depressed The model of cytokine therapy has been recently used to
patients and controls who are similar in body weight investigate the physiopathology of cytokine-induced
and/or adiposity, metabolic alterations in adipose tissue depression. Because of their immunomodulatory, antiviral
signaling might contribute to increases of IL-6 in depres- and antiproliferative properties, cytokines, principally
sion independent of immune cell production of this interferon (IFN)-a and IL-2, are currently used for the
proinflammatory cytokine. treatment of immune-mediated medical illnesses, including
cancer and viral infections (e.g., chronic hepatitis C,
3.4. From cytokines to depression AIDS). However, cytokine therapies are notorious for
causing neurobehavioral symptoms, including major
An emerging hypothesis generated from the findings of depression, in up to 50% of patients undergoing treatment
immune activation in depression was that cytokine abnor- with the cytokine, IFN-a (Musselman et al., 2001; Capuron
malities might contribute to depression. Indeed, there are et al., 2002). Similar rates of depression have been reported
many reasons to believe that the inflammatory changes in patients treated with the cytokine, IL-2 (Capuron et al.,
found in depressed patients, described above, may in part 2004).
contribute to the pathophysiology of the disorder. First, There are two distinct behavioral syndromes with differ-
medically ill patients, who exhibit evidence of immune acti- ent phenomenology and responsiveness to antidepressants
vation and/or inflammation secondary to tissue damage in patients who become depressed with cytokine therapies
and destruction, infection, autoimmunity or neoplastic dis- (Capuron et al., 2002). The mood and cognitive syndrome,
ease, exhibit high rates of depression. Second, cytokine characterized by the typical symptoms of depression such
therapies for infectious diseases and cancer are notorious as depressed mood, anxiety, irritability, memory and atten-
for causing behavioral alterations. Finally, there are many tional disturbance, develops usually between the first and
pathways known to be involved in the pathophysiology of third month of IFN-a therapy in vulnerable patients
depression that are influenced by cytokines, including (Musselman et al., 2001; Capuron et al., 2002; Capuron
neuroendocrine function, neurotransmitter function and et al., 2004). In contrast, the neurovegetative syndrome,
information processing. characterized by symptoms of fatigue, psychomotor
380 M.R. Irwin, A.H. Miller / Brain, Behavior, and Immunity 21 (2007) 374–383

slowing, anorexia and altered sleep patterns, develops ear- exaggerated release of ACTH and cortisol as compared
lier (within two weeks of IFN-a therapy) in a large propor- to responses in cancer patients who remained free of
tion of patients and persists at later stages of therapy depression (Capuron et al., 2003b). Moreover, a significant
(Capuron et al., 2002). In terms of their responsiveness to correlation was found between the degree of the initial
antidepressants, the mood and cognitive syndrome was ACTH and cortisol response to IFN-a and the develop-
found to be highly responsive to pretreatment with the ment of depression, anxiety and cognitive dysfunction after
antidepressant paroxetine (a selective serotonin inhibitor), 8 weeks of IFN-a treatment (Capuron et al., 2003b).
whereas the neurovegetative syndrome was antidepressant Another pathway by which cytokines may influence the
non-responsive (Capuron et al., 2002). Together, these data neuroendocrine system and thereby contribute to depres-
suggest that differential pathophysiological pathways may sion is through disruption of the functioning of the gluco-
be involved in the development of specific symptom dimen- corticoid receptor (GR) (Pariante et al., 1999). Patients
sions including mood/cognitive versus neurovegetative with major depression have reliably been shown to exhibit
symptoms in the context of cytokine system activation. alterations in GR function as manifested both in vivo (as
reflected by an abnormal dexamethasone suppression test
3.4.3. Pathways linking immunity to depression and/or dexamethasone-CRH test) and in vitro (as reflected
Given that proinflammatory cytokines are relatively by reduced sensitivity of peripheral blood mononuclear
large molecules that do not freely cross the blood brain cells to dexamethasone-induced inhibition of immune cell
barrier, research has focused on how cytokine signals reach function). Of specific relevance to the GR, inflammatory
the brain. At least three pathways have been invoked: (1) and immunoregulatory cytokines have been shown to alter
passage through leaky regions in the blood brain barrier, virtually every aspect of GR function including GR expres-
(2) active transport via cytokine-specific transport mole- sion, GR phosphorylation state, GR translocation, GR
cules and (3) activation of vagal afferent nerves which sig- protein-protein interactions and ultimately GR binding to
nal relevant brain nuclei such as the nucleus of the solitary DNA (Pace et al., 2007). The signal transduction pathways
tract (Raison et al., 2006). Once cytokine signals reach the by which cytokines affect GR function also have been
brain, there is a cytokine network within the brain that can described and include NFjB, p38 MAPK, JNK and
amplify and transpose relevant signals into those that inter- STAT5 (Pace et al., 2007). Finally, cytokines (e.g. IL-2
act with pathophysiologic pathways that are known to be and IL-4) have been shown to induce the b isoform of
involved in the development of depression. the GR which is inactive and thereby serves to compete
Central cytokines may contribute to the pathophysiol- for ligand and reduce activation of the GR a isoform, the
ogy of depression by their effects on neuroendocrine func- primary mediator of GR effects (Pace et al., 2007). Given
tion. For example, under normal conditions, inhibitory the role of glucocorticoids in regulating immune activation
elements such as glucocorticoids and anti-inflammatory and inflammation, cytokine-induced disruption of GR
cytokines limit the activation of the immune system to lev- function may lead to a feed-forward cascade in which
els that are appropriate for clearing the initial antigenic increasing inflammation leads to increased glucocorticoid
stimulus. However, it is becoming increasingly recognized resistance which in turn leads to reduced glucocorticoid-
that in certain conditions these inhibitory feedback loops mediated feedback inhibition of inflammatory responses
become impaired, allowing for chronic immune activation (Raison and Miller, 2003).
and the persistence of sickness symptoms that follow from Both in animal and humans, cytokines have been shown
this activation. Depression may correspond to one of these to have profound effects on the metabolism of brain neuro-
conditions where inhibitory feedback loops are altered and transmitters. In many studies, cytokines have been shown to
both neuroendocrine and immune systems become persis- interfere with 5HT metabolism and activity, a mechanism
tently activated (Raison and Miller, 2003). that may account for the depressogenic effects of cytokines.
A number of inflammatory and immunoregulatory cyto- Indeed, significant and consistent decreases in serum/plasma
kines have been found to have potent effects on the HPA concentrations of tryptophan, the primary precursor of
axis. One consistent finding is the capacity of the cytokines 5HT, have been reported in patients undergoing IL-2 and/
that mediate innate immune responses (e.g. IFN-a, IL-1, or IFN-a therapy (Capuron et al., 2002). At the experimen-
IL-6 and TNF-a) to increase the release of corticotrophin tal level, treatments with the cytokines, IL-1-b and TNF-a,
releasing hormone (CRH); hypersecretion of CRH is a reli- have been found to up-regulate significantly the expression
able finding in patients with major depression as mani- and activity of the 5HT transporter both in human cells
fested by increased cerebrospinal fluid concentrations of and cell lines, an effect which may contribute to reduced syn-
CRH and increased CRH mRNA and protein in the hypo- aptic availability of serotonin (Zhu et al., 2006).
thalamus of postmortem samples from depressed patients. Strong support for a role of 5HT alterations in cytokine-
Recent studies in humans have also indicated that cyto- induced depression is the involvement of cytokines in the
kine-induced activation of the HPA axis may represent a induction of the enzyme, indoleamine 2,3 dioxygenase
risk marker for depression (Capuron et al., 2003b). Among (IDO). IDO is induced by cytokines, especially IFN-
cancer patients who developed major depression during gamma, in a variety of immune cells including monocyte-
IFN-a therapy, the first injection of IFN-a induced an derived macrophage and microglia, upon immune system
M.R. Irwin, A.H. Miller / Brain, Behavior, and Immunity 21 (2007) 374–383 381

activation. IDO catalyzes the rate-limiting step of trypto- targeted at the pathways by which cytokines may contrib-
phan conversion into kynurenine and then quinolinic acid, ute to depression. Probably the most obvious target are the
thereby reducing the availability of tryptophan for conver- proinflammatory cytokines themselves, including the use of
sion into 5-HT, the primary mediator of serotonin effects in IL-1 receptor antagonist (e.g. anakinra) to target IL-1; the
the brain. Of relevance to depressed patients, TRP deple- soluble TNF receptor (e.g. etanercept) or antibodies to
tion in patients who became depressed during IFN-a ther- TNF (e.g. infliximab) to target TNF; and the anti-inflam-
apy was associated with marked increases in kynurenine, matory cytokine, IL-10, to target multiple inflammatory
suggesting that increased IDO activity contributed to the cytokines. Such cytokine ‘‘antagonists’’ may find both sys-
TRP decreases observed in depressed patients (Capuron temic usefulness as well as local applications such as the
et al., 2003a). Taken together, these data support the treatment of peripheral nerve pathological pain syndromes
notion that IDO may represent a key player in the patho- (Watkins et al., 2003). Of note, etanercept, infliximab and
physiology of cytokine-induced depression. anakinra are all available commercially for the treatment
Aside from effects on neurotransmitter and neuroendo- of rheumatoid arthritis, and therefore are available for pre-
crine function, recent data suggest that cytokines are able liminary analyses of efficacy in patients with altered mood
to alter higher cognitive brain functions including funda- status. Indeed, 12 weeks of etanercept administration led to
mental information processing. Indeed, in a recent brain improvements in depressive symptom severity in 618 psori-
functional magnetic resonance imaging (fMRI) study, asis patients (Tyring et al., 2006). Targeting cytokine sig-
patients with chronic hepatitis C (HCV) treated with naling pathways are also quite relevant, especially given
IFN-a for 12 weeks were found to exhibit significant acti- the increased emphasis on drug development in cancer
vation in the dorsal part of the anterior cingulate cortex and cardiovascular disease where relevant cytokine signal-
(Capuron et al., 2005), a brain region that has been ing pathways including NFjB and p38 MAPK are also
involved in conflict monitoring and cognitive control dur- implicated in the pathophysiology of these diseases. Other
ing cognitive tasks with high-demand. Interestingly, in promising approaches include the targeting of CRH, which
IFN-a treated HCV patients, activation of the cingulate as noted above is induced by proinflammatory cytokines
cortex highly correlated with errors made in a visuo-spatial and has been shown to cause many of the same symptoms
attention task administered during the fMRI session as sickness behavior when administered to laboratory ani-
(Capuron et al., 2005). These data suggest that cytokines mals. Several pharmaceutical companies (Stout et al., 2002)
(IFN-a) induce alterations in information processing (as are developing antidepressants targeting CRH, and novel
revealed by increased activity in the anterior cingulate cor- CRH antagonists. Other targets include inflammatory
tex) that may manifest by an increased sensitivity to pro- mediators such as the prostaglandins as well as the CNS
cessing conflicts and events perceived as potentially monoamines such as serotonin, norepinephrine and dopa-
threatening (Capuron et al., 2005); possibly reflecting a mine. Given the capacity of cytokine exposure to influence
so-called ‘‘danger’’ signal in the brain. Such changes in brain monoamines, therapies targeted at influencing mono-
brain activity and relevant cognitive processes may in turn amine neurotransmission (including selected antidepres-
impart an increased vulnerability to negative affects and sants as well inhibitors of IDO) might be especially
more generally to psychopathology (e.g., mood disorders). useful. Finally, because glucocorticoids serve to potently
Moving further ’’upstream’’, the most recent studies inhibit inflammatory signaling pathways, pharmacologic
examining the role of the immune system in depression agents that enhance glucocorticoid signaling and/or inhibit
have begun to explore the inflammatory signaling path- inflammatory signaling, including type 4 phospodiesterase
ways that may contribute to the pathophysiologies in neu- inhibitors, may be worthy of consideration (McKay and
rotransmitter function, neuroendocrine function and Cidlowski, 1999; Miller et al., 2002a; Pace et al., 2007).
neural circuitry described above. Specifically, studies have
identified NFjB signaling pathways as critical for the entry 5. Conclusions
of cytokine signals into the brain (Nadjar et al., 2005), and
p38 mitogen activated protein kinase has been found to There is strong evidence that depression involves altera-
play a key role in the effects of cytokines on serotonin tions in multiple aspects of immunity that may not only
transporter function as well as glucocorticoid receptor contribute to the development or exacerbation of a number
function (Zhu et al., 2006; Pace et al., 2007). of medical disorders but also may contribute to the patho-
physiology of the disease itself. Accordingly, aggressive
4. The future . . . 2007 and beyond management of depressive disorders in medically ill popu-
lations or individuals at risk for disease may improve dis-
4.1. Translational implications ease outcome or prevent disease development. On the
other hand, in light of data suggesting that immune
Based on the potential role of cytokines in the patho- processes may interact with the pathophysiologic pathways
physiology of depression, opportunities exist for ‘‘transla- known to contribute to depression, novel approaches to the
tional’’ research strategies that focus on the management treatment of depression may target relevant aspects of the
of sickness/depressive symptoms using novel therapies immune response. Taken together, the data provide
382 M.R. Irwin, A.H. Miller / Brain, Behavior, and Immunity 21 (2007) 374–383

compelling evidence that a psychoneuroimmunologic Circulating natural killer cell phenotypes in men and women with
frame of reference may have profound implications regard- major depression. Arch. Gen. Psychiatry 49, 388–395.
Fawzy, F.I., Canada, A.L., Fawzy, N.W., 2003. Malignant melanoma:
ing the consequences and treatment of depression. effects of a brief, structured psychiatric intervention on survival and
recurrence at 10-year follow-up. Arch. Gen. Psychiatry 60, 100–103.
Acknowledgments Frank, M.G., Hendricks, S.E., Johnson, D.R., Wieseler, J.L., Burke, W.J.,
1999. Antidepressants augment natural killer cell activity: in vivo and
This work was supported in part by Grants HL 079955, in vitro. Neuropsychobiology 39, 18–24.
Friedman, E.M., Irwin, M., 2001. Central CRH suppresses specific
AG 026364, CA 10014152, MH 19925, MH55253, M01 antibody responses: effects of beta-adrenoceptor antagonism and
RR00827, General Clinical Research Centers Program, adrenalectomy. Brain Behav. Immun. 15, 65–77.
and the Cousins Center for Psychoneuroimmunology. Herbert, T.B., Cohen, S., 1993. Depression and immunity—a meta-
The authors have no financial gain related to the out- analytic review. Psychol. Bull. 113, 472–486.
come of this research, and there are no potential conflicts Irwin, M., 2002. Effects of sleep and sleep loss on immunity and cytokines.
Brain Behav. Immun. 16, 503–512.
of interest. All authors have reviewed and approved the fi- Irwin, M., Brown, M., Patterson, T., Hauger, R., Mascovich, A., Grant,
nal version of this manuscript. I., 1991. Neuropeptide Y and natural killer cell activity: findings in
depression and Alzheimer caregiver stress. FASEB J. 5, 3100–3107.
References Irwin, M., Caldwell, C., Smith, T.L., Brown, S., Schuckit, M.A., Gillin,
J.C., 1990a. Major depressive disorder, alcoholism, and reduced
Andreoli, A.V., Keller, S.E., Rabaeus, M., Marin, P., Bartlett, J.A., natural killer cell cytotoxicity: role of severity of depressive symptoms
Taban, C., 1993. Depression and immunity: age, severity, and clinical and alcohol consumption. Arch. Gen. Psychiatry 47, 713–719.
course. Brain. Behav. Immun. 7, 279–292. Irwin, M., Clark, C., Kennedy, B., Gillin, J.C., Ziegler, M., 2003. Nocturnal
Anisman, H., Ravindran, A.V., Griffiths, J., Merali, Z., 1999. Endocrine catehcolamines and immune function in insomniacs, depressed patients,
and cytokine correlates of major depression and dysthymia with and control subjects. Brain Behav. Immun. 17, 365–372.
typical or atypical features. Mol. Psychiatry 4, 182–188. Irwin, M., Costlow, C., Williams, H., Artin, K.H., Chan, C.Y., Stinson,
Antoni, M.H., Lutgendorf, S.K., Cole, S.W., Dhabhar, F.S., Sephton, D.L., Levin, M.J., Hayward, A.R., Oxman, M.N., 1998. Cellular
S.E., McDonald, P.G., Stefanek, M., Sood, A.K., 2006. The influence immunity to varicella-zoster virus in patients with major depression. J.
of bio-behavioural factors on tumour biology: pathways and mecha- Infect. Dis. 178 (Suppl 1), S104–S108.
nisms. Nat. Rev. Cancer 6, 240–248. Irwin, M., Daniels, M., Smith, T.L., Bloom, E., Weiner, H., 1987a.
Capuron, L., Gumnick, J.F., Musselman, D.L., Lawson, D.H., Reem- Impaired natural killer cell activity during bereavement. Brain Behav.
snyder, A., Nemeroff, C.B., Miller, A.H., 2002. Neurobehavioral Immun. 1, 98–104.
effects of interferon-alpha in cancer patients: phenomenology and Irwin, M., Hauger, R., Jones, L., Provencio, M., Britten, K.T., 1990b.
paroxetine responsiveness of symptom dimensions. Neuropsychophar- Sympathetic nervous system mediates central corticotropin-releasing
macology 26, 643–652. factor induced suppression of natural killer cytotoxicity. J. Pharm.
Capuron, L., Neurauter, G., Musselman, D.L., Lawson, D.H., Nemeroff, Exp. Ther. 255, 101–107.
C.B., Fuchs, D., Miller, A.H., 2003a. Interferon-alpha-induced Irwin, M., Lacher, U., Caldwell, C., 1992. Depression and reduced natural
changes in tryptophan metabolism. Relationship to depression and killer cytotoxicity: a longitudinal study of depressed patients and
paroxetine treatment. Biol. Psychiatry 54, 906–914. control subjects. Psychol. Med. 22, 1045–1050.
Capuron, L., Pagnoni, G., Demetrashvili, M., Woolwine, B.J., Nemeroff, Irwin, M., McClintick, J., Costlow, C., Fortner, M., White, J., Gillin, J.C.,
C.B., Berns, G.S., Miller, A.H., 2005. Anterior cingulate activation 1996. Partial night sleep deprivation reduces natural killer and cellular
and error processing during interferon-alpha treatment. Biol. Psychi- immune responses in humans. FASEB J. 10, 643–653.
atry 58, 190–196. Irwin, M., Patterson, T., Smith, T.L., Caldwell, C., Brown, S.A., Gillin,
Capuron, L., Raison, C.L., Musselman, D.L., Lawson, D.H., Nemeroff, J.C., Grant, I., 1990c. Reduction of immune function in life stress and
C.B., Miller, A.H., 2003b. Association of exaggerated HPA axis depression. Biol. Psychiatry 27, 22–30.
response to the initial injection of interferon-alpha with development Irwin, M., Vale, W., Rivier, C., 1990d. Central corticotropin-releasing
of depression during interferon-alpha therapy. Am. J. Psychiatry 160, factor mediates the suppressive effect of stress on natural killer
1342–1345. cytotoxicity. Endocrinology 126, 2837–2844.
Capuron, L., Ravaud, A., Miller, A.H., Dantzer, R., 2004. Baseline mood Irwin, M.R., Vale, W., Britton, K.T., 1987b. Central corticotropin-
and psychosocial characteristics of patients developing depressive releasing factor suppresses natural killer cytotoxicity. Brain Behav.
symptoms during interleukin-2 and/or interferon-alpha cancer ther- Immun. 1, 81–87.
apy. Brain Behav. Immun. 18, 205–213. Irwin, M.R., Wang, M., Campomayor, C.O., Collado-Hidalgo, A., Cole, S.,
Cole, S.W., Naliboff, B.D., Kemeny, M.E., Griswold, M.P., Fahey, J.L., 2006. Sleep deprivation and activation of morning levels of cellular and
Zack, J.A., 2001. Impaired response to HAART in HIV-infected genomic markers of inflammation. Arch. Intern. Med. 166, 1756–1762.
individuals with high autonomic nervous system activity. Proc. Natl. Jung, W., Irwin, M., 1999. Reduction of natural killer cytotoxic activity in
Acad. Sci. USA 98, 12695–12700. major depression: Interaction between depression and cigarette smok-
Collado-Hidalgo, A., Bower, J.E., Ganz, P.A., Cole, S.W., Irwin, M.R., ing. Psychosom. Med. 61, 263–270.
2006. Inflammatory biomarkers for persistent fatigue in breast cancer Kaestner, F., Hettich, M., Peters, M., Sibrowski, W., Hetzel, G., Ponath,
survivors. Clin. Cancer Res. 12, 2759–2766. G., Arolt, V., Cassens, U., Rothermundt, M. 2005. Different activation
Cover, H., Irwin, M., 1994. Immunity and depression:insomnia,retarda- patterns of proinflammatory cytokines in melancholic and non-
tion, and reduction of natural killer cell activity. J. Behav. Med. 17, melancholic major depression are associated with HPA axis activity.
217–223. J Affect Disord.
Cruess, D.G., Douglas, S.D., Petitto, J.M., Have, T.T., Gettes, D., Dube, Kronfol, Z., House, J.D., 1985. Depression, hypothalamic-pituitary
B., Cary, M., Evans, D.L., 2005. Association of resolution of major adrenocortical activity and lymphocyte function. Psychopharmacol.
depression with increased natural killer cell activity among HIV- Bull. 21, 476–478.
seropositive women. Am. J. Psychiatry 162, 2125–2130. Kronfol, Z., House, J.D., Silva, J., Greden, J., Carroll, B.J., 1986.
Evans, D.L., Folds, J.D., Petitto, J.M., Golden, R.N., Pedersen, C.A., Depression, urinary free cortisol excretion, and lymphocyte function.
Corrigan, M., Gilmore, J.H., Silva, S.G., Quade, D., Ozer, H., 1992. Br. J. Psychiatry 148, 70–73.
M.R. Irwin, A.H. Miller / Brain, Behavior, and Immunity 21 (2007) 374–383 383

Lesperance, F., Frasure-Smith, N., Theroux, P., Irwin, M., 2004. The Raison, C., Miller, A.H., 2003. When not enough is too much: the role of
association between major depression and levels of soluble intercellular insufficient glucocorticoid signaling in the pathophysiology of stress-
adhesion molecule 1, interleukin-6, and C-reactive protein in patients related disorders. Am. J. Psychiatry. 160, 1554–1565.
with recent acute coronary syndromes. Am. J. Psychiatry 161, 271– Raison, C.L., Capuron, L., Miller, A.H., 2006. Cytokines sing the blues:
277. inflammation and the pathogenesis of depression. Trends Immunol.
Maes, M., Bosmans, E., Meltze, H.Y., Scharpe, S., Suy, E., 1993. 27, 24–31.
Interleukin-1 beta: a putative meidator of HPA axis hyperactivity in Ravindran, A.V., Griffiths, J., Merali, Z., Anisman, H., 1995.
major depression. Am. J. Psychiatry 150, 1189–1193. Lymphocyte subsets associated with major depression and dysthy-
Maes, M., Meltzer, H.Y., Stevens, W., Cosyns, P., Blockx, P., 1994. mia: modification by antidepressant treatment. Psychosom. Med. 57,
Multiple reciprocal relationships between in vivo cellular immunity 555–563.
and hypothalamic-pituitary-adrenal axis in depression. Psychol. Med. Sanders, V.M., Straub, R.H., 2002. Norepinephrine, the beta-adrenergic
24, 167–177. receptor, and immunity. Brain Behav. Immun. 16, 290–332.
Maes, M., Vandoolaeghe, E., Ranjan, R., Bosmans, E., Van Gastel, A., Schlatter, J., Ortuno, F., Cervera-Enguix, S., 2004. Lymphocyte subsets
Bergmans, R., Desnyder, R., 1996. Increased serum soluble CD8 or and lymphokine production in patients with melancholic versus
suppressor/cytotoxic antigen concentrations in depression: suppressive nonmelancholic depression. Psychiatry Res. 128, 259–265.
effects of glucocorticoids. Biol. Psychiatry 40, 1273–1281. Schleifer, S.J., Eckholdt, H.M., Cohen, J., Keller, S.E., 1993. Analysis of
McKay, L.I., Cidlowski, J.A., 1999. Molecular control of immune/ partial variance (APV) as a statistical approach to control day to day
inflammatory responses: interactions between nuclear factor-kappa variation in immune assays. Brain Behav. Immun. 7, 243–252.
B and steroid receptor-signaling pathways. Endocr. Rev. 20, 435– Schleifer, S.J., Keller, S.E., Bartlett, J.A., 1999. Depression and
459. immunity: clinical factors and therapeutic course. Psychiatry Res.
Miller, A.H., Vogt, G.J., Pearce, B.D., 2002a. The phosphodiesterase type 85, 63–69.
4 inhibitor, rolipram, enhances glucocorticoid receptor function. Schleifer, S.J., Keller, S.E., Bond, R.N., Cohen, J., Stein, M., 1989. Major
Neuropsychopharmocology 27, 939–948. depressive disorder and immunity: role of age, sex, severity, and
Miller, G.E., Cohen, S., Herbert, T.B., 1999. Pathways linking major hospitalization. Arch. Gen. Psychiatry 46, 81–87.
depression and immunity in ambulatory female patients. Psychosom. Schleifer, S.J., Keller, S.E., Czaja, S., 2006. Major depression and
Med. 61, 850–860. immunity in alcohol-dependent persons. Brain Behav. Immun. 20,
Miller, G.E., Stetler, C.A., Carney, R.M., Freedland, K.E., Banks, W.A., 80–91.
2002b. Clinical depression and inflammatory risk markers for coro- Seidel, A., Arolt, V., Hunstiger, M., Rink, L., Behnisch, A., Kirchner, H.,
nary heart disease. Am. J. Cardiol. 90, 1279–1283. 1995. Cytokine production and serum proteins in depression. Scan J.
Moynihan, J.A., Ader, R., Grota, L.J., Schachtman, T.R., Cohen, N., Immun. 41, 534–538.
1990. The effects of stress on the development of immunological Stein, M., Miller, A.H., Trestman, R.L., 1991. Depression, the
memory following low-dose antigen priming in mice. Brain Behav. immune system, and health and illness. Arch. Gen. Psychiatry
Immun. 4, 1–12. 48, 171–177.
Musselman, D.L., Miller, A.H., Porter, M.R., Manatunga, A.K., Gao, F., Stout, S.C., Owens, M.J., Nemeroff, C.B., 2002. Regulation of cortico-
Penna, S., Pearce, B.D., Landry, J., Glover, S., McDaniel, J.S., tropin-releasing factor neuronal systems and hypothalamic-pituitary-
Nemeroff, C.B., 2001. Higher than normal plasma interleukin-6 adrenal axis activity by stress and chronic antidepressant treatment. J.
concentrations in cancer patients with depression: preliminary find- Pharmacol. Exp. Ther. 300, 1085–1092.
ings. Am. J. Psychiatry 158, 1252–1257. Strausbaugh, H., Irwin, M., 1992. Central corticotropin releasing
Nadjar, A., Bluthe, R.M., May, M.J., Dantzer, R., Parnet, P., 2005. hormone reduces cellular immunity. Brain Behav. Immun. 6, 11–17.
Inactivation of the cerebral NFkappaB pathway inhibits interleukin- Thomas, A.J., Davis, S., Morris, C., Jackson, E., Harrison, R., O’Brien,
1beta-induced sickness behavior and c-Fos expression in various brain J.T., 2005. Increase in interleukin-1beta in late-life depression. Am. J.
nuclei. Neuropsychopharmacology 30, 1492–1499. Psychiatry 162, 175–177.
Nemeroff, C.B., Widerlov, E., Bissette, G., Walleus, H., Karlsson, I., Tyring, S., Gottlieb, A., Papp, K., Gordon, K., Leonardi, C., Wang, A.,
Eklund, K., Kilts, C.D., Loosen, P.T., Vale, W., 1984. Elevated Lalla, D., Woolley, M., Jahreis, A., Zitnik, R., Cella, D., Krishnan, R.,
concentrations of CSF corticotropin-releasing-factor-like immunore- 2006. Etanercept and clinical outcomes, fatigue, and depression in
activity in depressed patients. Science 226, 1342–1344. psoriasis: double-blind placebo-controlled randomised phase III trial.
Nieman, D.C., Nehlsen-Cannarella, S.L., Markoff, P.A., Balk-Lamberton, Lancet 367, 29–35.
A.J., Yang, H., Chritton, D.B.W., Lee, J.W., Arabatzis, K., 1990. The Vedhara, K., Cox, N.K., Wilcock, G.K., Perks, P., Hunt, M., Anderson,
effects of moderate exercise training on natural killer cells and acute S., Lightman, S.L., Shanks, N.M., 1999. Chronic stress in elderly
upper respiratory tract infections. Int. J. Sports Med. 11, 467–473. carers of dementia patients and antibody response to influenza
Pace, T.W., Hu, F., Miller, A.H., 2007. Cytokine-effects on glucocorticoid vaccination [see comments]. Lancet 353, 627–631.
receptor function: relevance to glucocorticoid resistance and the Watkins, L.R., Milligan, E.D., Maier, S.F., 2003. Glial proinflammatory
pathophysiology and treatment of major depression. Brain Behav. cytokines mediate exaggerated pain states: implications for clinical
Immun. 21, 9–19. pain. Adv. Exp. Med. Biol. 521, 1–21.
Pace, T.W., Mletzko, T.C., Alagbe, O., Musselman, D.L., Nemeroff, C.B., Yirmiya, R., Weidenfeld, J., Pollak, Y., Morag, M., Morag, A., Avitsur,
Miller, A.H., Heim, C.M., 2006. Increased stress-induced inflamma- R., Barak, O., Reichenberg, A., Cohen, E., Shavit, Y., Ovadia, H.,
tory responses in male patients with major depression and increased 1999. Cytokines, ‘‘depression due to a general medical condition,’’ and
early life stress. Am. J. Psychiatry 163, 1630–1633. antidepressant drugs. Adv. Exp. Med. Biol. 461, 283–316.
Page, G.G., Ben-Eliyahu, S., Liebskind, J.C., 1994. The role of LGL/NK Zautra, A.J., Yocum, D.C., Villanueva, I., Smith, B., Davis, M.C., Attrep,
Cells in surgery induced promotion of metastasis and its attenuation J., Irwin, M., 2004. Immune activation and depression in women with
by morphine. Brain Behav. Immun. 8, 241–250. rheumatoid arthritis. J. Rheumatol. 31, 457–463.
Pariante, C.M., Pearce, B.D., Pisell, T.L., Sanchez, C.I., Po, C., Su, C., Zhu, C.B., Blakely, R.D., Hewlett, W.A., 2006. The proinflammatory
Miller, A.H., 1999. The proinflammatory cytokine, interleukin-1alpha, cytokines interleukin-1beta and tumor necrosis factor-alpha activate
reduces glucocorticoid receptor translocation and function. Endocri- serotonin transporters. Neuropsychopharmacology 31, 2121–2131.
nology 140, 4359–4366. Zorrilla, E.P., Luborsky, L., McKay, J.R., Rosenthal, R., Houldin, A.,
Pike, J.L., Irwin, M.R., 2006. Dissociation of inflammatory markers and Tax, A., McCorkle, R., Seligman, D.A., Schmidt, K., 2001. The
natural killer cell activity in major depressive disorder. Brain Behav. relationship of depression and stressors to immunological assays: a
Immun. 20, 169–174. meta-analytic review. Brain Behav. Immun. 15, 199–226.

Вам также может понравиться