p-ISSN: 1454-7848
e-ISSN: 2068-7176

IMPLICATIONS OF THE INFLAMMATORY PROCESSES IN MAJOR DEPRESSIVE EPISODE

Abstract

Abstract There are an increasing number of evidences that suggests that the immune system and inflammatory processes can contribute to MDD pathogenesis in a significant proportion. In this article, we tried to review the current evidences that are suggesting that inflammatory processes contribute to the development of MDD by direct actions on the brain as well as by effects on secondary pathways that are linking the brain to other system of the organism. We consider that more and more evidences are linking the immunomodulatory interventions to mood variation and this may hold the promise of a new approach in MDD treatment, especially in patients with elevations of the inflammatory biomarkers. These interventions refer to cytokine and cyclooxygenase antagonists, but also to agents that impact inflammatory transcription factors or signaling cascades.

Until now, the studies have only been able to establish a bidirectional interaction between the brain and body’s immune system but these findings are more relevant in the way that an individual is more likely to develop an episode of Major Depressive Disorder (MDD) in response to both stress and inflammatory disease than other individual with normal immune response. Also, it has been established that the psychosocial stressors are among the most replicated and reliable risk factors for MDD (1) most likely because the psychosocial stress activates the inflammatory response and this is the major link between stress and MDD. For example, the exposure to the Trier Social Stress Test (TSST – a public speaking and mental arithmetic stressor) was associated with a significant increase in DNA binding of the inflammatory transcription factor (NFkβ) in peripheral blood mononuclear cells (PBMCs), whereas merely watching others undergoing the TSST had no such effect (2). Others studies suggested that psychosocial stressors may result in enhanced inflammatory responses in individuals with early life adversity no matter if these people are currently depressed or not depressed in that moment (3, 4). It is possible that an early life stress like trauma or maltreatment can represent a powerful risk factor for adult MDD by inducing a long term increases in inflammatory signaling because the inflammatory biomarkers are elevated if these people are depressed, but this status is maintained even in the not depressed state (5).
S o m e a r t i c l e s i n d i c a t e a l s o t h a t t h e parasympathetic nervous system is involved in immune regulation and has potent anti-inflammatory effects (6). The inhibitory effects on the inflammatory response, referred to as the ‘cholinergic anti-inflammatory pathway’, have been proved to be mediated by vagal release of acetylcholine that in turn activates the alpha7 subunit of the nicotinic acetylcholine receptor which can regulate both cytokine transcription and translation (6). Moreover, some studies suggest that T cells may contribute to this inhibitory cholinergic reflex (7). In addition, the reduced heart rate variability which is a reflection of reduced parasympathetic tone, has been associated with increased inflammatory biomarkers including IL—6 and CRP in medically healthy individuals as well as patients with MDD and heart disease (8, 9). On the other hand, the stress might activate the inflammatory response and the evidences most strongly support the role of the central autonomic nervous system (10).
T h e r e l a t i o n s h i p b e t w e e n M D D a n d inflammation on an experimental pathophysiological model, was initially demonstrated by the fact that the cytokine administration induces depressive symptoms. For example, chronic administration of the inflammatory cytokine, interferon (IFN)-CT, has also been found to induce depressive symptoms, with as many as 30—50% of IFNα—treated patients meeting symptom criteria for MDD (11). Moreover, supporting the similarity between the MDD associated with IFNα and depression in other populations is the fact that IFNα—induced MDD can be prevented and/or treated by conventional antidepressants therapy (12, 13, 14, 15).
Recent evidences suggest that the inflammatory response itself can contribute to the various measures of suppressed immunity observed in MDD, including direct effects of inflammatory cytokines on signaling through the T cell receptor (16, 17, 18, 19, 20). Many articles and meta-analyses confirm that peripheral blood elevations in the cytokines like IL-6, tumor necrosis factor (TNF-α), and the acute phase reactant, C reactive protein (CRP), chemokines and cellular adhesion molecules in peripheral blood as well as increased stress-induced nuclear factor- kβ (NFkβ) are among the most reliable biomarkers of increased inflammation in MDD patients (21, 22, 23, 24). Moreover, many studies have found significant a s s o c i a t i o n s b e t w e e n b l o o d c o n c e n t r a t i o n s o f inflammatory factors and the severity of depressive symptoms but the proofs are somehow inconsistent if we refer to the fact that increased inflammatory markers in MDD patients return to control levels after successful antidepressant treatment (25 ,26, 27, 28).
These findings suggest that failure to respond to antidepressant treatment should be associated with ongoing elevations in inflammatory biomarkers, and indeed several studies support this theory (29, 30). Out of these data we can appreciate that an elevation in inflammatory markers prior to treatment might predict poor response, suggesting a relationship between inflammation and treatment resistance (29, 30, 31, 32). This can be explained by the fact that the administration of cytokines modifies the serotonin and dopamine level but also the glutamate level (28). Moreover, the serotonin reuptake inhibitors (SSRI) can prevent or treat depressive symptoms during chronic exposure to IFNα patients with infectious diseases and cancer provides strong evidence that serotonin pathways are involved in cytokine effects on behavior (12, 33). Further implicating of the alterations in serotonin metabolism in cytokine induced behavioral changes, are studies showing that IFNα associated increases in cerebrospinal fluid (CSF) concentrations of IL-6 are inversely correlated with the serotonin metabolite, 5-hydroxyindoleacetic acid (5—HIAA), which, in turn, negatively correlate with IFNα that induced depression severity (34). Also, cytokines were proved to have a significant impact on dopamine pathways. Studies have found altered blood flow and metabolic activity in basal ganglia nuclei during exposure to inflammatory stimuli (35, 36, 37), and in studies in non- human primates, reduced CSF concentrations of the dopamine metabolite, homovanillic acid (HVA), were associated with depressive-like huddling behavior secondary to chronic IFNα administration (38). This is h a p p e n i n g b e c a u s e t h e e n z y m e i n d o l e a m i n e 2,3—dioxygenase (IDO) is activated by various cytokines alone or in combination (39, 40). IDO catabolizes tryptophan, the primary amino acid precursor of serotonin, into kynurenine. Depletion of tryptophan provides protection against a variety of pathogens, but also inhibits effector T cell responses and thereby contributes to immune tolerance (41). IDO increases the production of kynurenine and its metabolites for immune- induced depression (42, 43).
Another pathway that can influence monoamine metabolism is the cytokine signaling pathway mitogen—activated protein kinase (MAPK). It was demonstrated that stimulation of p38 MAPK pathways increases the expression and function of the serotonin transporter (SERT), which in animal studies has been shown to increase depressive-like behavior in response to inflammatory stimuli (44). In humans, increased phosphorylation of p38 MAPK following the first injection of IFNα was associated with the subsequent development of IFNα-induced depression and fatigue (45). In addition to effects on serotonin metabolism, MAPK pathways have also been found to influence the dopamine transporter (DAT) (46). Moreover, the inflammatory mediators are known to profoundly impact glutamatergic functioning in the CNS because cytokines have been shown to decrease the expression of glutamate transporters on glial cells and to increase the release of glutamate from astrocytes (47, 48, 49). This astrocyte- derived glutamate has preferential access to extra synaptic NMDA receptors, which can mediate excitotoxicity and lead to decreased production of trophic factors including brain—derived neurotrophic factor (BDNF) (50, 51).
The hypothalamic—pituitary—adrenal (HPA) axis anomalies and their relationship with mood are one of the most reproducible findings in patients with MDD because these patients have been found to exhibit increased concentrations of the HPA axis hormones (like ACTH and cortisol) and an increase in CSF measures of t h e H P A a x i s r e g u l a t o r y n e u r o p e p t i d e , corticotrophin—releasing hormone (CRH) (52, 53). Moreover, the studies in patients have proven that the acute ACTH and cortisol response to the first injection of IFNα (presumably due to activation of CRH pathways) is powerful and relates with the subsequent development of depressive symptoms during IFNα therapy in patients with cancer but the acute administration of IFNα, chronic IFNα administration is associated with flattening of the d i u r n a l c u r v e a n d i n c r e a s e d e v e n i n g c o r t i s o l concentrations, both of which correlate with the development of depression and fatigue (54, 55). This finding is important because the flattening of the diurnal cortisol rhythm has been seen in many medical disorders associated with inflammation including cardiovascular disease and cancer, where it has been associated with a worse outcome in these diseases (55). For example, previous studies have shown that the flattened cortisol rhythm is associated with non-suppression of cortisol by dexamethasone (DEX) in the DEX suppression test (DST) (56). DST non-suppression is a common finding in major depression both in vivo and in vitro in peripheral blood mononuclear cells (PBMCs) (52, 53, 57). DEX non- suppression, also referred to as glucocorticoid resistance, has additionally been correlated with stimulated production of IL-18 by PBMCs of patients with MDD (58). The inflammatory cytokines can disrupt glucocorticoid receptor (GR) function while decreasing GR expression (59). A similar protein-protein interaction between the GR and NFkβ in the nucleus has also been described (60). On the other hand, IL-Ia and IL-IB have been shown both in vitro and in Vivo to inhibit GR translocation from the cytoplasm to the nucleus through activation of p38 MAPK (61, 62). Moreover, the stress induced alterations in GR translocation leading to glucocorticoid resistance in laboratory mice has been found to be mediated by ILel (using IL-1 KO mice), who fail to exhibit impaired GR translocation following social disruption stress (61). So, chronic exposure to inflammatory glucocorticoids can lead to decreased GR expression as well as increased expression of GRB (GRB expression has been proven to be increased in patients with inflammatory disorders including patients with bronchial asthma and rheumatoid arthritis), a GR isoform which has a distinct hormone binding domain (that is unable to bind known glucocorticoids) and a unique pattern of gene regulation (59, 63).
N e u r o g e n e s i s i s s t i m u l a t e d b y m a n y antidepressant medications (by increasing the BDNF level) and it is important in the MDD recovery and in the relapses prevention (64). The data have showed that the stress induces a decrease in neurogenesis and on the expression of relevant nerve growth factors, including BDNF (which support neurogenesis). This can be reversed by administration of the IL-1 receptor antagonist or transplantation of 1L—1Ra secreting neural precursor cells into the hippocampus or the use of IL-1 receptor knock—out (KO) mice (65, 66, 67). In patients, the treatment with IFNα has been associated with reduced levels of periphera1 BDNF, which are known to correlate well with BDNF availability in the CNS (68).
In patients under IFNα therapy for cancer or C hepatitis (by using positron emission tomography (PET SCAN)), was found a marked increase in glucose metabolic activity in the basal ganglia which correlated with symptoms of fatigue (69, 70). These increases were found also in patients with Parkinson’s disease and they are believed to reflect increased oscillatory burst activity in relevant basal ganglia nuclei secondary to DA depletion (71,72).
Another brain region that has been found to be influenced by cytokine administration and inflammatory stimuli is the dACC that plays an important role in error detection and conflict monitoring (73). Patients with hepatitis C treated with IFNα were found to have significantly higher activation of the dACC using fMRI and a task of visuospatial attention (12, 74). A strong correlation was found in this study between activation of the dACC in IFNα-treated patients and the number of errors made during the task. The error rate was quite low for the task, and no such correlation was found in control subjects. In this context, the patients with high-trait anxiety have also been shown to exhibit increased dACC activation during fMRI in the context of low error rates (75). Moreover, increased activation of the dACC after a psychosocial stressor (TSST) was correlated with the degree of activation of peripheral blood IL—6, suggesting a role for stress-induced inflammation in the findings (76). Moreover, the dACC plays a role in processing the social pain, also (77). Given the connection of the dACC with downstream autonomic nervous system arousal pathways, these investigators have further hypothesized that the dACC may serve as a “neural alarm system” that can both detect and respond (with arousal and distress) to threatening environmental stimuli in the social domain (77). In this context, the cytokines may sensitize the responsivity of the dACC, thereby contributing to the anxiety, arousal and alarm that often accompany chronic exposure to inflammatory stimuli such as IFNα. From an evolutionary perspective, this heightened dACC activity (and heightened sensitivity to social threat) may sub serve the survival priority of vigilance against attack in an animal that is otherwise vulnerable due to infection or wounding (78). In conclusion, the administration of an inflammatory stimulus has been associated with activation of the sub-genual ACC, the target of deep brain stimulation strategies in patients with treatment resistant depression (79).
Because the immune system interacts with nearly every system in the brain and body, many therapeutically strategies can be putatively work for MDD treatment.
Probably the best studied anti-inflammatory strategy for the treatment of depression is the targeting of inflammatory signaling pathways. The simplest approach in this regard can be the administration of pharmacologic agents that inhibit cyclo-oxygenase (COX – the enzyme that converts arachidonic acid into prostaglandin) because the prostaglandins have been demonstrated to be increased in depression and are known to play an important role in the inflammatory response including the mediation of fever and sensitivity to pain. Moreover, inhibition of COX—1 and COX—2 selectively and in combination have been shown in laboratory animals to inhibit depressive like behavior following administration of the inflammatory stimulus without influencing cytokine responses (80, 81). Still, there are some controversy as to whether COX—1 versus COX-2 is the most relevant target. Generally, the indomethacin and ibuprofen (nonselective COX inhibitors), have some proofs that showed an efficacy in animal models (81). In patient’s studies, the add-on of the acetylsalicylic acid (which blocks COX-1 and COXeZ) to fluoxetine led to increased remission rates in an open label study of 24 depressed patients previously nonresponsive to fluoxetine alone (82). Another study of 20 medically healthy depressed patients who received the COXeZ inhibitor, celecoxib, in combination with reboxetine showed greater symptomatic improvement compared to patients randomized to reboxetine plus placebo (83). But, both studies involved small sample sizes, and in the celecoxib study, 50% or more of the sample in each group dropped out before conclusion of the study (83).
Another therapeutically add-on strategy could target the cytokine because they are probably one of the most obvious targets for treating the impact of peripheral inflammation on the brain. Some case reports indicated that several of the currently available biologics therapies (monoclonal antibodies, soluble cytokine receptors or other antagonists which combine cytokine receptors with other fusion proteins that target cytokines were effective in reducing symptoms of depression and fatigue as well as quality of life (84).
Another possible therapeutically strategy could include indoleamine 2,3—dioxygenase (IDO) as a potential target for the unique contributions of the immune system to depression is evidenced by studies that have administered an IDO antagonist or used IDO KO mice in the context of immune activation or infection. Thus, there are great hopes regarding the development of more potent IDO inhibitors (85). In some studies of the mice treatment with the IDO antagonist, 1—methyl tryptophan (I—M’I’), abrogates the impact of LPS as well as infection with bacillus Calmettee Guerin (BCG), an attenuated form of mycobacterium bovis, on depressive like behavior including increased immobility in the PST and TST (86, 87). Targeted deletion of KAT II (the enzyme that converts KYN to KA) has also been shown to increase cognitive performance in association with an increase in the amplitude of long—term potentiation in vitro, while reducing extracellular KA as measured by hippocampal in vivo micro dialysis (88). Moreover, given the elaboration of the kynurenine pathway as well as data indicating the role of kynurenic acid and quinolinic acid in neuropsychiatric disease, especially neurodegenerative disorders, few compounds which interfere with multiple steps in the kynurenine pathway have been identified in clouding inhibitors of 3-hydroxyanthranilic acid oxygenase which converts 3ehydroxyanthranilic acid to quinolinic acid (42, 89).
A l s o , w e m u s t m e n t i o n a s a p o s s i b l e therapeutically target the NFkβ because it plays a big role in the inflammatory response by transmitting inflammatory signals from the periphery to the brain, where it has been proven to mediate inflammation induced inhibition of neurogenesis (90, 91). Out of all the compounds that have been shown to inhibit NFkβ, a special interest has been recently paid to natural compounds such as curcumin (a derivative of the curry spice, turmeric), which has been shown to decrease NFkβ activation in PBMCs of patients with pancreatic cancer (92). Another natural compound is d—tocopherol (a form of vitamin E) which has been shown to block NFkβ and the associated neuro-inflammatory and behavioral response to LPS (93), and resveratrol (found in the skin of red grapes) which has been shown to inhibit LPS- induced induction of inflammatory cytokines, chemokines and inducible nitric oxide synthase (iNOS) in murine microglia and astrocytes in association with inhibition of NFKfi (94).
In the end, we must not forget to mention that the activation of the parasympathetic nervous system is involved in immune regulation and has potent anti- inflammatory effects (95) and because of that most behavioral interventions that have shown antidepressant properties, including psychotherapy, exercise and weight loss, also tend to reduce peripheral inflammatory biomarkers. Recently, meditation practices have also shown evidence of anti-inflammatory activity. The practice of a secularized compassion meditation technique has been reported to reduce inflammatory responses (i.e. plasma IL-6) to a standardized laboratory stress or in healthy young adults and to reduce resting state levels of CRP in highly traumatized adolescents in state custody (96, 97, 98). These studies also observed behavioral effects of the training that might be expected to either reduce or prevent depressive pathology. Also, the mindfulness based stress reduction has been reported to reduce feelings of loneliness in older individuals and to down-regulate Nfkβ associated gene expression profiles (99). Similar changes in the balance of pro to anti- inflammatory transcription activity as assessed by gene expression analyses have been observed in caregivers of dementia patients trained in yoga meditation (100).

References
1.Kendler KS, Gardner CO: Dependent stressful life events and prior depressive episodes in the prediction of major depression: the problem of causal interence in psychiatric epidemiology. Arch Gen Psychiatry
2010;67:1120—1127.
2.Bierhaus A et al: A mechanism converting psychosocial stress into mononuclear cell activation. Proc Natl Acad Sci USA 2003;100:1920- 1925.
3.Face TW, Mletzko TC, Alagbe O, et a1: Increased stress-induced inflammatory responses in male patients with major depression and increased early life stress. Am I Psychiatry 2006;163:1630-1633.
4.Carpenter LL, et al: Association between plasma 1L-6 response to a c u t e s t r e s s a n d e a r l y – l i f e a d v e r s i t y i n h e a l t h y a d u l t s . Neuropsychopharmacology 2010;35:2617-2623
5.Brown DW et al: Adverse childhood experiences and the risk of premature mortality. Am J Prev Med 2009;37:389-396.
6.Tracey KI et al: Reflex control of immunity. Nat Rev Irnmunol 2009;9:418—428.
7.O’Mahony C et al: Loss of vagal anti inflammatory effect: in vivo visualization and adoptive transfer. Am J Physiol Regul Integr Comp Physiol 2009;297:R1118—R1126.
8.Frasure-Smith N, Lesperance F, Irwin MR, Talajic M: The relationships among heart rate variability, inflammatory markers and depression in coronary heart disease patients. Brain Behav Immun
2009;23:1140-1147.
9.Fischer IE, lhayer IF: Heart rate variability, overnight urinary norepinephrine and C-reactive protein: evidence for the cholinergic anti- inflammatory pathway in healthy human adults. I Intern Med
2009;265:439—447.
10.Iohnson ID, Campisi I, Sharkey CM, et al: Catecholarnines mediate stress—induced increases in peripheral and central inflammatory cytokines. Neuroscience 2005;135:1295—1307.
11.Raison CL, Demetrashvili M, Capuron L, Miller AH: Neuropsychiatric side effects of interferon-alpha: recognition and management. CNS Drugs 2005;19:1-19.
12.Musselman DL, Lawson DH, Gumnick JF, et a1: Paroxetine for the prevention of depression induced by high-dose interferon alfa.
N Engl J Med 2001;344: 96 1—966.
13.Raison CL, Woolwine BI, Demetrashvili MF, et al: Paroxetine for prevention of depressive symptoms induced by interferon-alpha and r i b a v i r i n f o r h e p a – t i t i s C . A l i m e n t P h a r m a c o l T h e r 2007;251l163—1174.
14.Kraus MR, Sehafer A, Schottker K, et al: Therapy of interferon- induced depression in chronic hepatitis C with citalopram: a randomised, double-blind, p1aceho-controlled study. Gut 2008;57:531—536.
15.Hauser P, Khosla I, Aurora H, et al: A prospective study of the incidence and open—label treatment of interferon induced major depressive disorder in patients with hepatitis C. MoI Psychiatry 2002;7:942—947.
16.Irwin MR, Miller AH: Depressive disorders and immunity: 20 years of progress and discovery. Brain Behav Immun 2007;21:374—383.
17.Blume J, Douglas SD, Evans DL: Immune suppression and immune activation in depression. Brain Behav Immun 201l;25:221-229.
18.Clark L Vagenas P, Panesar M, Cope AP: What does tumour necrosis factor excess do to the immune system long term? Ann Rheum Dis 2005;64(suppl 4):iv70-iv76.
19.Lotrich FE, Ferrell RE, Rabinovitz M, Pollock BG: Risk for depression during interferon-alpha treatment is affected by the serotonin transporter polymorphism. Biol Psychiatry 2009;65:344-348.
20.Iuengling FD, Ebert D, Gut O, et al: Prefrontal cortical hypometabolism during low dose interferon alpha treatment. Psychopharmacology 2000;152:383-389.
21.Zorilla F, Lulborsky L, McKay J, Roesenthal R, et al: The relationship of depression and stressors to immunological assays: a meta-analytic review Brain Behav Immun 2001;15:199—226.
22.Dowlati Y, Herrinann N, Swardfager W, et al: A meta-analysis of cytokines in major depression. Biol Psychiatry 2010;67:446—457.
23.Howren MB, Lamkin DM, Suls J: Associations of depression with C- reactive protein, IL-I, and IL-6: a meta~analysis. Psychosom Med 2009;71:171— 186.
24.Haroon E, Raison CL, Miller AI-I: Psychoneuroimmunulogy meets neuropsychopharmacology: translational implications of the impact of i n fl a m m a t i o n o n b e h a v i o r. N e u r o p s y c h o p h a r m a c o l o g y 2012;37:1377162.
25.Bower IE, Ganz PA, Aziz N, Fahey IL: Fatigue and prointlammatory cytokine activity in breast cancer survivors. Psychosorn Med 2002;64:604—611.
26.Miller GE et al: Clinical depression and inflammatory risk markers for coronary heart disease. Am J Cardiol1012002;90:1279—1283.
27.Alesci S, Martinez PE, Kelkar S, et al: Major depression is associated with significant diurnal elevations in plasma interleukin~6 levels, a shift of its circadian rhythm, and loss of physiological complexity in its secretion: clinical implications. I Clin Endocrinol Metah 2005;90:2522- 2530.
28.Motivala SI, Sarfatti A, Olmos L, lrwin MR: Inflammatory markers and sleep disturbance in major depression. Psychosom Med 2005;67:187—194.
29.Lanquillon S, Krieg IC, Bening-Abu-Shach U et al: Cytokine production and treatment response in major depressive disorder. Neuropsychopharmacology 2000;22:370—379.
30.Fitzgerald P, O’Brien SM, Scully P et al: Cutaneous glucocorticoid receptor sensitivity and pro—inflammatory cytokine levels in antidepressant-resistant depression. Psychol Med 2006;36:37—43.
31.Benedetti F, Lucca A, Brambilla F et al: Interleukine-6 serum levels correlate with response to antidepressant sleep deprivation and sleep phase advance. Prog Neuro-Psychopharmacol Biol Psychiatry 2002;26:1167—1170.
32.Harley I, Luty S, Carter I, Mulder R et al: Elevated C-reactive protein in depression: a predictor of good long, term outcome with an tid ep r es s an ts an d p o o r o u tco me w ith p s y ch o th er ap y. I Psychopharrnacol 2010;24:625-626.
33.Raison CL, Woolwine BI, Demetrashvili MF, et al: Paroxetine for prevention of depressive symptoms induced by interferon alpha and ribavirin for hepatitis C. Aliment Pharm Ther 2007;25:1163-1174.
34.Raison CL, Borisov AS, Majer M, et al: Activation of central nervous system inflammatory pathways by interferon alpha: relationship to monoamines and depression. Biol Psychiatry 2009;65:296-303.
35.Brydnn L, Harrison NA, Walker C, Steptoe A. Critchiey HD: Peripheral inflammation is associated with altered substantia nigra activity and psychomotor slowing in humans. Biol Psychiatry 2008;63:1022—1029
36.Iuengling FD, Ebert D, Gut O, et al: Prefrontal cortical hypometabolism during low dose interferon alpha treatment. Psychopharmacology 2000;152:383-389.
37.Capuron L, Pagnoni G, Demetrashvili MF, et al: Basal ganglia hypermetabolism and symptoms of fatigue during interferon alpha therapy. Neuropsychopharmacology 2007;32:2384—2392.
38.Felger JC, Alagbe 0, Ho F, et al: Effects of interferon-alpha on rhesus monkeys: a nonhuman primate model of cytokine—induced depression. Biol Psychiatry 2007;62:1324—1333.
39.Pemberton LA, Kerr SI, Smythe G, Brew BI: Quinolinic acid production by macrophages stimulated with IFN-gamma, TNF-alpha, and IFN alpha. J Interferon Cytoldne Res 1997;17:589—595.
40.Fujigaki H, Saito K, Fujigaki S, et al: The signal transducer and activator of transcription lalpha and interferon regulatory factor 1 are not essential for the induction of indoleamine 2,3-dioxygenase by lipopolysaccharide: involvement of p38 mitogen-activated protein kinase and nuclear factor-kappaB pathways, and synergistic effect of several proinflammatory cytokines. J Biochem 2006; 139:655-662.
41.Huang L, Baban B, Iohnson BA 3rd, Mellor AL: Dendritic cells, indoleamine 2,3 dioxygenase and acquired immune privilege, 1111 Rev Immun012010;29:133—155.
42.Schwarcz R, Pellicciari R: Manipulation of brain kynurenines: glial targets, neuronal effects, and clinical opportunities.
J Pharmacol Exp Ther 2002;303:1—10.
43.Raison CL, Dantzer R, Kelley KW et al: CSF concentrations of brain tryptophan and kynurenines during immune stimulation with IFNealpha: relationship to CNS immune responses and depression. Mol Psychiatry 2010;15:393-403.
44.Zhu CB, Blakely RD, Hewlett WA: The proinflarnmatory cytokines interleukin-1heta and tumor necrosis factorialpha activate serotonin transporters. Neuropsychopharmacology 2006;31:2121—2131.
45.Felger IC, Alagbe 0, Pace TW et al: Early activation of p38 mitogen activated protein kinase is associated with interferon alpha-induced depression and fatigue. Brain Behav Immun 201 1:25:1094—1098.
46.Moron IA, Zakharova I, Ferrer IV et al: Mitogen—activated protein kinase regulates dopamine transporter surface expression and dopamine transport capacity. I Neurosci 2003;23:8480—8488.
47.Volterra A, Meldolesi I: Astrocytes, from brain glue to communication elements: the revolution continues. Nat Rev Neurosci 2005;6:626-640.
48.Tilleux S, IIermans E: Neuroinflamation and regulation of glial glutamate uptake in neurological disorders. J Neurosci Res
2007;85:2059-2070.
49.Ida T, Hara M, Nalramura Y, Kozaki S, Tsunoda S, Ihara H: Cytokine- induced enhancement of calcium-dependent glutamate release from astrocytes mediated by nitric oxide. Neurosci Lett 2005:32:232-236.
50.Hardingham GE, Fukunaga Y, Bading H; Extrasynaptic NMDARs oppose synaptic NMDARs by triggering CREE shut off and cell death pathways. Nat Neurosci 2002;5:405-414.
51.Haydon PG, Carmignoto G: Astrocyte control of synaptic transmission and neurovascular coupling. Physiol Rev 2006;86:1009- 1031.
52.Pariante CM, Miller AH: Glucocorticoid receptors in major depression: relevance to pathophysiology and treatment. Biol Psychiatry 2001;49:391-404.
53.Pariante CM, Lightman SL: The HPA axis in major depression: classical theories and new developments. Trends Neurosci 2008;31:464—468.
54.Raison CL, Borisov AS, Woolwine BI, Massung B, Vogt G, Miller AII: Interferon-alpha effects on diurnal hypothalamic-pituitary-adrenal axis activity: relationship with proinflammatory cytokines and behavior. Mol Psychiatry 2010;15:535-547.
55.Sephton SE, Sapolsky RM, Kraemer HC, Spiegel D: Diurnal cortisol rhythm as a predictor of breast cancer survival. J Natl Cancer Inst 2000;92:994-1000.
56.Spiegel D, Glese-Davis J, Taylor CB, Kraemer H: Stress sensitivity in metastatic breast cancer: analysis of hypothalamic-pituitary-adrenal axis function. Psychoneuroendocrinology 2006;31:1231-1244.
57.Pariante CM: Glucocorticoid receptor function in vitro in patients with major depression. Stress 2004;7:209-219.
58.Maes M, Bosmans E, Meltzer HY, Scharpe S, Suy E: Interleukin—1 beta: a putative mediator of HPA axis hyperactivity in major depression? Am J Psychiatry 1993;150:1189—1193.
59.Pace TW, Hu F, Miller AH: Cytokine effects on glucocorticoid receptor function: relevance to glucocorticoid resistance and the pathophysiology and treatment of major depression. Brain Behav lrnmun 2007;21:9— 19.
60.Smoak KA, Cidlowski IA: Mechanisms of glucocorticoid receptor signaling during inflammation. Mech Age Dev 2004;125:697—706.
61.Engler H, Bailey MT, Engler A, Stiner-Iones LM, Quan N, Sheridan IF: Interleukinel receptor type 1-deficient mice fail to develop social s t r e s s – a s s o c i a t e d g l u c o c o r t i c o i d r e s i s t a n c e i n t h e s p l e e n . Psychoneuroendocrinology 2008;33: 108-117.
62.Pariante CM, Pearce BD, Pisell TL, et al: The proinflammatory cytokine, interleukin lalpha, reduces glucocorticoid receptor translocation and function. Endocrinology 1999;140:4359—4366.
63.Kino T, Manoli I, Kelkar S, Wang Y, Su YA, Chrousos GP: Glucocorticoid receptor (GR) beta has intrinsic, GRalpha-independent t r a n s c r i p t i o n a l a c t i v i t y. B i o c h e m B i o p h y s R e s C o m m u n 2009;381:671—675.
64.Duman RS, Monteggia LM: A neurotrophic model for stress-related mood disorders. Biol Psychiatry 2006;59:1l16-1127.
65.Barrientos RM, Sprunger DB, Campeau S, et a1: Brain-derived neurotrophic factor mRNA downregulation produced by social isolation is blocked by intrahippocampal interleukine-l receptor antagonist. Neuroscience 2003;121:847-853.
66.Ben Menachem-Zidon O, Goshen I, Kreisel T, et al: Intrahippocampal transplantation of transgenic neural precursor cells overexpressing interleukin-1 receptor antagonist blocks chronic isolationeinduced impairment in memory and neurogenesis. Neuropsychopharmacology 2008;315:2251—2262.
67.Koo IW, Duman RS: IL-lbcta is an essential mediator of the antineurogenic and anhedonic effects of stress. Proc Natl Acad Sci USA 2008;105:751—756.
68.Kenis G, Prickaerts J, van Os J, et al: Depressive symptoms following interferon—alpha therapy: mediated by immune induced reductions in brain, derived neurotrophic factor? Int J Neuropsychopharrnacol 2011;14:247—253.
69.Capuron L, Pagnoni G, Demetrashvili MF, et al: Basal ganglia hypermetabolism and symptoms of fatigue during interferonalpha therapy. Neuropsychopharmacology 2007;32:2384—2392.
70.Iuengling FD, Ebert D, Cut O, et al: Prefrontal cortical hypornetabolism during low-dose interferon alpha treatment. Psychopharmacology (Berl) 2000;152:383-389.
71.Wichmann T, DeLong MR: Oscillations in the basal ganglia. Nature 1999;400:621—622.
72.Feigin A, Fukuda M, Dhawan V, et al: Metabolic correlates of levodopa response in Parkinson’s disease. Neuroleogy 2001;57:2083- 2088.
73.Carter CS, Braver TS, Barch DM, Botvinick MM, Noll D, Cohen ID: Anterior Cingulate cortex, error detection, and the online monitoring of performance. Science 1998;280:747—749.
74.Capuron L, Pagnoni G, Demetrashvili M, et al: Anterior cingulate activation and error processing during interferon-alpha treatment. Biol Psychiatry 2005;58:190-196.
75.Paulus MP, Peinstein IS, Simmons A, Stein MB: Anterior Cingulate activation in high trait anxious subjects is related to altered error processing during decision making. Biol Psychiatry 2004;55:1179-1187.
76.Slavich GM, Way BM, Eisenberger NI, Taylor SE: Neural sensitivity to social rejection is associated with inflammatory responses to social stress. Proc Natl Acad Sci USA 2010;107:14817—l4822.
77.Eisenberger Ni, Lieberman MD: Why rejection hurts: a common neural alarm system for physical and social pain. Trends Cogn Sci 2004;8:294-300.
78.Miller AH: Mechanisms of cytokine-induced behavioral changes: psychoneuroimmunology at the translational interface. Brain Behav Immun 2009;23:149-l58.
79.Lozano AM, Mayberg IIS, Giacobbe P, Harnani C, Craddock RC, Kennedy SH: Subcallosal Cingulate gyrus deep brain stimulation for treatment—resistant depression. Biol Psychiatry 2008,64:461-467.
80.Lima SN, de Paiva VN, Fernandes MM, Soncini R, Andrade CA, Giusti-Paiva A: Prostaglandins mediate depressive-like behaviour induced by endotoxin in mice. Behav Brain Res 2010;215:146-151.
81.Cunningham C, Teeling IL, Newman TA, Perry VH: The effect of non-steroidal anti-inflammatory agents on behaveoural changes and cytokine production following systemic inflammation: implications for a role of COX—1. Brain Behav Immun 2010;24:409-419.
82.Mendlewicz I, Kriwin P, Oswald P, Souery D, Alboni S, Brunello N: Shortened onset of action of antidepressants in major depression using acetylsalicylic acid augmentation: a pilot open-label study. Int Clin
Psychopharmacol 2006;21:227-231.
83.Muller N, Schwarz MI, Dehning S, et al: The cyclooxygenase-2 inhibitor celecoxib has therapeutic effects in major depression: results of a double—blind, randomized, placebo controlled, add-on pilot study to reboxetine. Mol Psychiatry 2006;11:680—684.
84.Soczynska IK, Kennedy SH, Goldstein BI, Lachowski A, Woldeyohannes HO, McIntyre R8: The effect of tumor necrosis factor antagonists on mood and mental health-associated quality of life: novel hypothesis-driven treatments for bipolar depression? Neurotoxicology 2009;30:497-52l.
85.Di Pucchio ‘1’, Danese S, De Cristofaro R, Rutella S: Inhibitors of indoleamine 2,3-dioxygenase: a review of novel patented lead compounds. Expert Opin Ther Pat 2010;20:229—250.
86.O’Connor IC, Lawson MA, Andre C, Moreau M, Lestage J, Castanon N, Kelley KW, Dantzer R: Lipopolysaccliaride-induced depressive-like behavior is mediated by indoleamine 2,3-di0xygenase activation in mice. Mol Psychiatry 2009;14:511-522.
87.O’Connor IC, Lawson MA, Andre C, et al: induction of IDO by bacille Calmette-Guerin is responsible for development of murine depressive- like behavior. J Immunol 2009;182:3202—3212.
88.Potter MC, Elmer GI, Bergeron R, et al: Reduction of endogenous kynurenic acid formation enhances extracellular glutamate, h i p p o c a m p a l p l a s t i c i t y , a n d c o g n i t i v e b e h a v i o r . Neuropsychopharmacology 2010;35:1734—1742.
89.Stone TW: Kynurenic acid antagonists and kynurenine pathway inhibitors, Expert Opin Investig Drugs 2001;10:633-645.
90.Koo IW, Russo SI, Ferguson D, Nestler EI, Duman RS: Nuclear factor-kappaB is a critical mediator of stress-impaired neurogenesis and depressive behavior. Proc Natl Acad Sci USA 2010;107:2669-2674.
91.Nadjar A, Bluthe RM, May MI, Dantzer R, Parnet P: Inactivation of the cerebral NFkappaB pathway inhibits interleukin-1beta-induced sickness behavior and c-Fos expression in various brain nuclei. Neuropsychopharmacology 2005;30:1492—1499.
92.Dhillon N, Aggarwal BB, Newman RA, et al: Phase II trial ofcurcumin in patients with advanced pancreatic cancer. Clin Cancer Res 2008,14:4491-4499.
93.Godbout IP, Berg BM, Krzyszton C, Johnson RW: Alpha-tocopherol attenuates NFkappaB activation and pro—inflarnmatory cytokine production in brain and improves recovery from lipopolysaccharide- induced sickness behavior. I Neuroimmunol 2005;169:97—105.
94.Lu X, Ma L, Ruan L, et al: Resveratrol differentially modulates i n fl a m m a t o r y r e s p o n s e s o f m i c r o g l i a a n d a s t r o c y t e s . J NeuroinflaJnmation 2010;7:46.
95.Tracey KI: Reflex control of immunity. Nat Rev Irnmunol 2009;9:418—428.
96.Pace TW, Negi LT, Adamo DD, et al: Effect of compassion meditation on neuroendocrine, innate immune and behavioral responses to psychosocial stress. Psychoneuroendocrinology 2009;34:87-98.
97.Pace TW, Negi LT, Sivilli TI, at al: Innate immune, neuroendocrinic and behavioral responses to psychosocial stress do not predict s u b s e q u e n t c o m p a s s i o n m e d i t a t i o n p r a c t i c e t i m e . Psychoneuroendocrinology 2010;35:310—315.
98.Face TW, Negi LT, Dodson-Lavelle B, et al: Engagement with cognitively-based compassion training is associated with reduced salivary C—reactive protein from before to after training in foster care program adolescents. Psychoneuroendocrinology 2012, E-pub ahead of print.
99.Creswell JD, Irwin MR. Burklund LJ, et al: Mindfulness-based stress reduction training reduces loneliness and proinflammatory gene expression in older adults: a small randomized controlled trial. Brain Behav Immun 2012;26:1095— l10l.
100.Black DS, Cole SW, Irwin MR, et al: Yogic meditation reverses NF- kappaB and IRF-related transcriptome dynamics in leukocytes of family d e m e n t i a c a r e – g i v e r s i n a r a n d o m i z e d c o n t r o l l e d t r i a l . Psychoneuroendocrinology 2012, Epub ahead of print.

***