Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Feb 1.
Published in final edited form as: Expert Rev Neurother. 2017 Nov 27;18(2):139–152. doi: 10.1080/14737175.2018.1407242

Immune-based strategies for mood disorders: facts and challenges

Gabriela D Colpo 1, Marion Leboyer 2, Robert Dantzer 3, Mahukar H Trivedi 4, Antonio L Teixeira 1
PMCID: PMC5819337  NIHMSID: NIHMS941664  PMID: 29179585

Abstract

Introduction

Inflammation seems to play a role in the pathophysiology of mood disorders, including major depressive disorder (MDD) and bipolar disorder (BD). In the last years several studies have shown increased levels of inflammatory and/or immune markers in patients with mood disorders. Accordingly, the immune system has become a target of interest for the development of biomarkers and therapeutics for mood disorders.

Areas covered

Here, we review the evidence showing low-grade inflammation in mood disorders and the studies evaluating immune-based strategies for the treatment of these conditions.

Expert Commentary

Clinical trials with non-steroidal anti-inflammatory drugs, polyunsaturated acids, N-acetylcysteine, anti-cytokines, physical activity and probiotics have provided promising results in terms of antidepressant efficacy in patients with MDD and BD. Regarding stem cells, only studies with animal models have been performed so far with interesting pre-clinical results. Due to the preliminary nature of the results, most of the clinical studies need to be replicated and/or confirmed in larger clinical settings, embracing the highly heterogeneous pathophysiology of mood disorders.

Keywords: Mood disorders, Major depressive disorder, Bipolar disorder, Inflammation, Neuroinflammation, Cytokines, Anti-inflammatory drugs, Immunomodulation

1. Introduction

Mood disorders including major depressive disorder (MDD) and bipolar disorder (BD) consist of a constellation of symptoms involving emotional, cognitive, and behavioral domains. They show high heritability relative to other common medical conditions but remain ‘idiopathic’ with respect to etiology [1].

The lifetime prevalence rates for MDD are 2–7%, and up to 16–20% suffer from milder forms of the illness. For BD the life-time prevalence rates are estimated at 1.0% for bipolar disorder I (BD-I), 1.1% for bipolar disorder II (BD-II) and 2.4% for subthreshold BD (2.4% total) [2, 3].

Besides relatively high prevalence rates, mood disorders have been recognized by the World Health Organization (WHO) as a major cause of disability worldwide [4, 5]. The WHO ranks MDD as the most important global cause of “years of life lived with disability” for all age groups, and projects that in 2030, MDD will rank first in global disease burden as measured in disability-adjusted life years [6]. Mood disorders are also associated with elevated mortality rates, especially due to suicide [7] and cardiovascular diseases [8]. The high magnitude of these global public health problems partly reflects the limited efficacy of the currently available therapies for mood disorders.

At least one-third of patients have treatment-resistant depression defined as lack of response to two or more antidepressants [9, 10, 11]. For these patients, novel antidepressant strategies are highly needed. However, development of drugs for the central nervous system (CNS) diseases has one of the lowest success rates [12]. A critical need is to identify safe and more effective treatments for mood symptoms by targeting receptors and/or signaling pathways beyond the monoamine systems [13, 14]. In addition, taking into account the complex interplay between individual variability in clinical phenotypes is important to develop and study treatments that match with the specific pathophysiological process, for example, immunological treatments for immune-related depressions and bipolar illnesses or circuit modulation for those with identified brain circuit dysfunctions. In this scenario, drug repurposing has been considered a cost-effective and reduced-risk strategy for developing new drugs for the CNS diseases [15].

Over the past decade, a compelling body of evidence has emerged to suggest the role of inflammation in the pathophysiology of mood disorders [16, 17, 18, 19]. These results have fostered the study of immune mechanisms and pathways as potential targets for pharmacological intervention for mood disorders. The purpose of this manuscript is to provide a comprehensive review and a commentary on the immune/inflammatory changes in mood disorders and immune-based treatments for MDD and BD.

2. Immune dysfunction in mood disorders

2.1 Major depression

Several studies have investigated the immune and/or inflammatory status of patients with MDD. One of the most consistent findings has been the observation of high levels of peripheral pro-inflammatory markers, notably C reactive protein (CRP), interleukin-6 (IL-6), tumor necrosis factor (TNF- α), and interleukin-1 receptor antagonist (IL-1ra) in patients with MDD [20, 21, 22, 23, 24]. Interestingly, higher CRP levels have been associated with increased risk of suicide in patients with MDD [25, 26].

Few studies investigated the central component (cerebrospinal fluid, CSF; and/or brain) of inflammation in patients with MDD. Medication-free patients with severe depression had higher concentrations of IL-1β, but lower IL-6 and no change in TNF-α in the CSF in comparison with healthy controls [27]. Low CSF concentrations of IL-6 and IL-6R were reported in medicated geriatric MDD patients [28]. Another study did not find any evidence of altered CSF levels of IL-6 in medication-free MDD patients [29].

In post-mortem studies, Dean et al showed that TNF-α receptors, TNFR1and TNFR2, had different expression in the cortex of patients with MDD when compared with controls [30]. A recent study showed differences in activation of inflammatory genes in the hippocampus of patients with MDD when compared with controls [31]. In 2014, Torres-Platas et al. provided the first evidence of increased microglial activation in post-mortem brain samples from middle-aged people with depression who committed suicide [32]. Positron emission tomography (PET) studies have also found evidence for increased translocator protein binding - interpreted as a marker of microglial activation - in the prefrontal cortex and anterior cingulate cortex of patients with MDD [33].

Microglial cells are the main resident immune cells of the brain. Microglia contains multimolecular complexes called inflammasomes. Inflammasomes function as intracellular sensors for infectious agents or cellular stress-signals. Once sensing endogenous danger signals (e.g. adenosine triphosphate or ATP), inflammasome can induce the release of the cytokines IL-1β and IL-18, and pyroptosis (caspase 1-dependent programmed cell death), contributing to the onset and/or progression of the inflammatory response [34]. Microglia also release other factors, such as glutamate, contributing even more to neuroinflammation [35]. Theoretically, a crosstalk between peripheral immune cells and microglia can potentiate inflammation both in the periphery and in the brain [36]. Lately, different brain cells as perivascular macrophages, oligodendrocytes and astrocytes have been identified as immune cells [37]. In psychiatric disorders, including MDD, one of the proposed mechanisms for this crosstalk involves the disruption of the blood-brain barrier (BBB) integrity with the endothelial cells increasing their permeability to pro-inflammatory mediators from the blood [38]. Another mechanism proposes that peripheral cytokines activate primary afferent nerves, such as the vagus nerve. This signal reaches first the nucleus tractus solitarius and subsequently the rest of the brain [39]. There is also a cellular pathway through which pro-inflammatory cytokines, mainly TNF-α, are able to stimulate microglia to produce the chemokine monocyte chemoattractant protein-1 (MCP-1/CCL3) which is responsible for the recruitment of circulating monocytes into the brain [40].

It is worth mentioning that these mechanisms have been proposed mainly after pre-clinical studies in which peripheral immune activation produces and/or exacerbates depressive-like behaviors in animal models, and these behaviors are prevented or reversed by different anti-inflammatory strategies [41].

Immune alterations observed in MDD patients may be associated with other inflammatory or immune-based diseases. There is a greater prevalence of autoimmune diseases in patients with depressive disorders than in the general population [42, 43]. Moreover, individuals with depressive disorders have almost twice the risk of developing cardiovascular diseases associated with low-grade inflammation, such as atherosclerosis [44, 45, 46]. Depression has also been associated with other conditions characterized by low-grade inflammation, including obesity, metabolic syndrome, and type II diabetes [47].

Together, these data suggest that MDD can be associated with a sustained low-grade systemic inflammation. However, in accordance with the phenotypic heterogeneity of MDD, a pattern of low-grade inflammation is not present in all patients with MDD. Some authors propose its occurrence in at least one third of MDD cases [48]. Others regard melancholic depression as a disorder of HPA axis activity [49], while atypical depression would be a more pro-inflammatory condition [50]. In addition, some studies have shown that the association between depressive states and inflammation differ according to the type of depressive symptoms endorsed. Accordingly, somatic or neurovegetative symptoms of depression (fatigue, sleep disturbances, poor appetite) are more associated with inflammation than emotional/cognitive symptoms (depressed mood, worthlessness, anhedonia, poor concentration) [51, 52].

2.2 Bipolar disorder

As in MDD, the majority of studies investigating the role of inflammation in BD assessed peripheral levels (i.e. plasma or serum) of cytokines, while only a few studies evaluated CSF cytokine levels in BD. CSF levels may be more relevant as they reflect better CNS levels and, therefore, any ongoing neuroinflammatory process. Studies evaluating CSF concentration of inflammatory markers, such as IL-1β, IL-8 and YKL-40 (marker of endothelial dysfunction), reported increased levels of these molecules in euthymic BD patients compared to healthy controls [53, 54, 55]. Regarding blood levels, most studies report higher peripheral levels of IL-6, IL-6R, IL-2R, IL-1β and TNF-α during depressive and acute manic episodes compared with healthy controls [56, 57, 58, 59, 60, 61, 62, 63, 64, 65]. Cross-sectional studies also observed increased CRP levels in BD patients during acute mania and/or a depressive phase compared with controls [64, 66, 67, 68, 69]. Accordingly, cytokine levels may vary depending on mood state. A recent systemic review corroborated the view that pro-inflammatory cytokines are increased, while anti-inflammatory cytokines are reduced in BD patients, mainly during manic and depressive phases, when compared to the controls. These peripheral changes tend to disappear during euthymia, indicating that inflammation may be associated with acute mood episodes of BD [19].

Few studies have measured inflammatory markers in post-mortem specimens. Rao et al observed higher protein and mRNA levels of IL-1β, IL-1R, and astroglial and microglial markers in the frontal cortex of BD patients compared with controls [70]. Other study showed differences in the pattern of activation of immune/inflammatory genes in the hippocampus of patients with BD and MDD when compared with control subjects [31]. It is highly possible that heightened levels of inflammatory mediators in the periphery might be translated to microglia, and vice-versa. In 2015, Patel and Frey suggested a model of BBB dysfunction in BD. In this model, the increase of oxidative stress and inflammation associated with BD might lead to BBB dysfunction and, therefore, decreased CNS protection with increased influx of peripheral mediators, triggering microglial activation, promoting more inflammation and neuronal damage [38]. However, direct evidence of microglial activation in BD is very limited. In a PET study with [(11)C]-(R)-PK11195, Haarman et al. observed a significant increase in microglial activation in the right hippocampus of patients with BD I compared with controls [71]. Interestingly, the same authors found a positive relation between microglial activation and neuronal integrity in vivo, suggesting that microglia’s role in BD is complex, and may also involve neuroprotection [72].

Differences in the profile of peripheral immune cells were reported in BD. For instance, Pandey et al. found that the mRNA expression of IL-1β, IL-6 and TNFα, as well as their receptors, IL-1R1, IL-1ra and TNFR1 was significantly increased in the lymphocytes from BD patients when compared with controls [73]. Our group showed that patients with BD I present a lower proportion of T lymphocytes, notably cytotoxic T cells and T regulatory cells, when compared with controls [74]. Alterations in intracellular pathways associated with pro-inflammatory status have also been described in peripheral blood mononuclear cells from BD patients [75].

Taken together, these results indicate that BD is associated with a pro-inflammatory state. This immune dysfunction may be an important link between mood disorders and other comorbidities. BD is strongly associated with auto-immune diseases, including Guillain-Barré syndrome, autoimmune hepatitis, multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, psoriasis and autoimmune thyroiditis [76, 77]. Moreover, patients with BD exhibit increased rates of obesity and metabolic syndrome, conditions associated with low-grade inflammation [78].

3. Immune-based strategies

3.1 Non-steroidal anti-inflammatory drugs

Non-steroidal anti-inflammatory drugs are well-known as a broad class of anti-inflammatory drugs that irreversibly inhibits cyclooxygenase-1 (COX-1) and cyclooxygenase -2 (COX-2), thereby decreasing prostaglandin and thromboxane levels, and the levels of pro-inflammatory cytokines, such as TNF-α and IL-6 [79]. Among them, celecoxib and acetyl-salicylic acid (ASA) have been studied in mood disorders.

Celecoxib has primarily been studied as an adjunctive therapy to conventional strategies for mood disorders. In three randomized clinical trials (RCTs) with MDD patients, celecoxib (200 mg 2×/day) was found to improve depressive symptoms as add-on therapy to, respectively, reboxetine 4 mg/day, fluoxetine 40 mg/day, and sertraline 200 mg/day (n = 40 for each RCT) [80, 81, 82]. A study involving patients with BD (depressive or mixed phase) compared the addition of celecoxib 200 mg 2×/day or placebo to standard treatment for 6 weeks, but no significant difference between groups was observed beyond the first week [83]. As celecoxib may increase the risk of cardiovascular diseases, some advocates that celecoxib should be avoided in the treatment of mood disorders [84].

Acetylsalicylic acid (ASA) is 50- to 100-fold more potent in inhibiting platelet COX-1 than monocyte COX-2 activity [85]. In addition, ASA has been found to increase the levels of lipoxins. Lipoxins are endogenous anti-inflammatory pro-resolving molecules that play a vital role in reducing tissue injury and chronic inflammation [86]. Mendlewicz et al. examined the effect of add-on ASA (160 mg/day) to conventional antidepressant pharmacotherapy in 24 patients with MDD proven to be non-responsive to 4 week treatment with SSRI. The combined administration of SSRI and ASA was associated with a response rate of 52.4%. Remission was achieved in 43% of the total sample and 82% of the responder sample. In the responder group, a significant improvement was observed within the first week of treatment and this benefit persisted through day 28 [87]. A retrospective study of 5,556 men aged 69 to 87 in different medical conditions, showed that the discontinuation of ASA resulted in elevation of depression scores [88].

In a large pharmaco-epidemiological study, Stolk et al. evaluated whether NSAIDs or glucocorticoids would improve BD symptoms in 5,145 patients receiving lithium. The subjects receiving low-dose (≤80 mg/day) ASA were 17% less likely to have a medication event (change in the type or number of psychotropic medications prescribed or increase in the psychotropic drug dose), a finding that remained significant after adjusting for age, sex, chronic disease score and healthcare utilization [89].

Taken together, the evidence of ASA as an effective therapeutic option for MDD and BD has still to be established, however, observational studies are promising and further investigation is currently underway. A clinical trial has just completed the investigation of ASA as an adjunctive therapy for bipolar depression (NCT01429272), and a clinical trial will start to investigate ASA for MDD (NCT03152409).

3.2 Minocycline

Minocycline is a tetracycline antibiotic that exerts anti-inflammatory, anti-oxidant, anti-glutamatergic and neuro-protective effects [90, 91]. For instance, high doses of minocycline reduce IL-1β levels in the brain of mice with traumatic brain injury [92]. Also, minocycline inhibits COX-2 expression and reduces prostaglandin E2 (PGE2) levels in microglial cell culture [93]. Minocycline readily crosses the BBB and attenuates inflammation associated with microglial activation [94]. Minocycline may be a potential candidate to adjunctive treatment for MDD and BD associated with microglial activation.

Miyaoka et al. used minocycline as an adjuvant to antidepressant medication (fluvoxamine, paroxetine or sertraline) in 25 patients with psychotic depression in a 6-week open-label study. Patients using minocycline had significant improvement in depressive and psychotic symptoms. No serious adverse events were observed [95]. Clinical trials are currently underway to investigate minocycline as an adjunctive therapy for unipolar (NCT01574742) and bipolar depression (NCT01429272, NCT01514422, NCT01403662).

3.3 Cytokine-inhibitors

TNF-α is produced by various immune cellular types, including activated macrophages, microglia and T-cells. TNF-α is a key cytokine to induce mood symptoms in the context of sickness behavior [96]. Sickness behavior is a term used to describe significant changes in subjective experience and behavior that occur in physically ill patients [96].

Currently, four molecules with anti-TNF-α activity are approved by the Food and Drug Administration (FDA 2012): adalimumab, a fully human monoclonal antibody; infliximab, a chimeric monoclonal antibody; etanercept, a soluble receptor construct; and certolizumab pegol, another monoclonal antibody. Etanercept and infliximab have been investigated for their efficacy in treating mood disorders.

Infliximab has significant anti-depressant effects in patients being treated for inflammatory conditions [97, 98, 99]. Kappelmann et al. performed a systematic review and meta-analysis of antidepressant activity of anti-cytokine clinical trials for chronic inflammatory conditions such as rheumatoid arthritis and psoriasis in which depressive symptoms were measured as a secondary outcome. The results showed robust improvement in depressive symptoms after treatment with cytokines inhibitors. The antidepressant effect was associated with the severity of depressive symptoms at baseline but not with the improvement of the primary disease [100].

Only one RCT to date has investigated the efficacy of a monoclonal antibody using depression as a primary outcome. Raison et al. studied the effect of three intravenous infliximab doses (5 mg/kg) (week 0, 2 weeks and 6 weeks) in a 12-week study in 60 patients moderately resistant to previous antidepressant therapy. Infliximab had an antidepressant effect only in individuals who had elevated levels of inflammatory markers, i.e. TNF-α and CRP, at baseline [101]. However, the post hoc nature of this result limits its significance.

A potential limiting factor of anti-TNF-α strategies is the significant risk of infection associated with them. There is an increased risk of infection for all anti-inflammatory strategies, and the infectious risk associated with anti-TNF-α agents is particularly concerning [102, 103, 104].

3.4 Polyunsaturated fatty acids and diet

Omega-3 polyunsaturated fatty acids (PUFAs) are a dietary fatty acid that cannot be endogenously produced by humans [105]. PUFAs exhibit anti-inflammatory effects by competing with arachidonic acid for COX enzymes, thereby decreasing PGE2 levels and pro-inflammatory cytokine production [106]. Omega-3 therapy lowers the inflammatory status in healthy middle-aged and old adults [107], and improves anxiety symptoms (but not depressive symptoms) in healthy young adults [108].

Meta-analyses comprising more than 15 RCTs evaluated the effectiveness of omega-3 PUFAs supplementation in adults with MDD. Most of them found that omega-3 supplementation was beneficial in adult patients with MDD, and but the effect was strongly dependent on the eicosapentaenoic acid content of nutritional regimens [109, 110]. Omega-3 PUFAs have also been shown to increase treatment efficacy when used as adjunctive to standard therapy for MDD [111, 112, 113].

There are five RCTs assessing the antidepressant effects of adjunctive omega-3 in patients with BD [114, 115, 116, 117, 118]. Two studies reported a significant reduction in depressive symptom severity compared with placebo [114, 117]. The other three studies found no significant difference in the reduction of depressive symptom severity [115, 116, 118]. Chiu et al. evaluated the effects of omega-3 in acutely manic inpatients with BD in a four-week RCT and found no difference compared with placebo [119].

The interest in diet intervention has gone beyond PUFAs. A recent RCT evaluated an adjunctive dietary intervention for the treatment of moderate to severe MDD. The diet was a modified Mediterranean diet that was delivered by a clinical dietitian. In comparison with the non-intervention group, the dietary support group showed significantly greater improvement of depressive symptoms between the baseline and 12 weeks [120]. A meta-analysis evaluated the possible association of fruit and vegetable intake with the risk of depression in the general population. Ten studies involving 227,852 participants for fruit intake and eight studies involving 218,699 participants for vegetable intake were analyzed. Both fruit 0.86 (RR) (CI: 0.81–0.91, p<0.001) and vegetable 0.89 (RR) (CI: 0.83–0.94, p<0.001) intake was significantly associated with decreased risk of depression [121]. These results suggest that dietary improvement may provide an effective and accessible treatment strategy for depression.

3.5 N-acetylcysteine

N-acetylcysteine (NAC) has been identified as a multi-target molecule, being considered primarily an antioxidant. NAC also has glutamatergic and anti-inflammatory properties. The ability of NAC to decrease neuroinflammation may be through inhibition of microglia. As mentioned before, microglial cells are phagocytic cells that can be activated by cytokines and in turn produce more inflammatory mediators, increase oxidative stress, and stimulate neurotoxicity [122]. NAC can inhibit cytokine and oxidative species production by macrophages and microglia [123]. This effect is likely to be through both stimulation of glutathione production and regulation of cystine/glutamate antiporters, with the result of reducing oxidative stress and glutamate excitotoxicity [124].

A 12-week RCT study with MDD patients reported that NAC was not superior to placebo [125]. In BD, there were two trials assessing mainly the antidepressant effect of NAC. Berk et al. studied 75 patients with BD under depressed, mixed or manic episodes in the last six months. Subjects received NAC 1,000 mg 2×/day or placebo in addition to the conventional therapy for 24 weeks. Adjunctive treatment with NAC decreased depressive symptoms in comparison with the placebo group [126]. In addition, Michael Berk’s group conducted a study evaluating adjunctive NAC for maintenance therapy in BD. During the first eight-weeks of the open-label phase of the trial, NAC significantly reduced depressive symptoms. However, in the double-blind phase, there were no significant differences between NAC and placebo groups [127]. Together these results indicate that adjunctive NAC may be useful for bipolar depression, but further studies are necessary to confirm this hypothesis.

3.6 Physical activity

It is well-known that regular physical activity reduces the risk of chronic metabolic and cardiovascular disease [128]. More recently, the ability of physical activity to improve mood symptoms in subjects with and without mood disorders has been assessed, with encouraging results [129, 130]. For instance, our group showed a positive effect of muscle strengthening and aerobic intervention on depressive symptoms in community-dwelling elderly women [130].

The antidepressant action of exercise has been proposed to derive from its effects on multiple pathways, including neurogenesis with increased expression of neurotrophic factors [131]. Exercise can also reduce systemic inflammation and oxidative stress [132]. The anti-inflammatory effects of regular exercise may be mediated by the reduction in visceral fat mass, increased production and release of anti-inflammatory cytokines from skeletal muscle and reduced expression of Toll-like receptors on monocytes and macrophages, with subsequent inhibition of downstream responses, such as the production of pro-inflammatory cytokines [133]. Exercise also modulates stress reactivity through effects on the HPA axis activity and cortisol production [131].

An early open trial reported that exercise was beneficial for depression and anxiety in patients with several psychiatric conditions, but only depression improvement was maintained at one-year follow-up [134]. Other open trials have also found adjunctive exercise beneficial for MDD [135, 136, 137].

In a recent meta-analysis involving 6 studies on 198 adults with depression, Shuck et al. found that exercise improved quality of life [138]. Adjusting for publication bias, the same authors showed that exercise has a large and significant antidepressant effect in people with depression, claiming that exercise is an evidence-based treatment for depression [129].

In BD, a small retrospective chart review found that depression and anxiety improved significantly among bipolar inpatients (depressed or manic) who participated voluntarily in an adjunctive exercise program [139]. Also, exercise augmentation was found beneficial in a small open trial [135] and in a small RCT [140] with MDD and BD depression subjects.

3.7 Probiotics

There is an extensive bidirectional communication between the gastrointestinal tract and the CNS, commonly referred as the gut–brain axis. The pathways by which this bidirectional communication takes place remain to be elucidated and involve a combination of neural, endocrine, and immune factors [141, 142].

Probiotics have traditionally been used to ‘normalize’ physiological dysfunctions such as gastrointestinal symptoms [143]. In recent years, studies have shown that probiotics are capable of changing behavior through the modulation of the gut-brain axis. Ultimately, treatment with probiotics may improve behavioral symptoms by acting on monoaminergic systems (e.g. increasing serotonin availability) and/or decreasing levels of systemic inflammatory markers [144].

Probiotics decreased the circulating levels of the pro-inflammatory cytokines IL-1β [145, 146], IL-6 and TNF-α [145], as well as microglial activation markers in animal models [147]. Chronic administration of probiotics protected rats from depression-like behavior induced by maternal deprivation, a condition associated with increased peripheral inflammation [148]. More recently, authors observed reduced Lactobacilli in stressed mice. Restoring intestinal Lactobacillus levels was sufficient to improve metabolic and behavioral changes [149].

Additionally, rats that received a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) for two weeks had significantly reduced anxiety-like behavior [150]. Interestingly, the same probiotic formulation was tested in humans. Volunteers who took it for 30 days had significant improvement on psychometric measures of anxiety, hostility and depression [150]. In another study, consumption of Lactobacillus was associated with a significant decrease of anxiety in patients with chronic fatigue syndrome [151].

Five clinical trials comprising 183 cases and 182 controls showed that probiotics significantly decreased depressive symptoms in patients with MDD and healthy subjects [152].

3.8 Stem cells

Stem cells, especially, mesenchymal stem cells (MSCs) are a type of multipotent stem cells that can be differentiated into several cell lineages [153, 154]. MSCs have the ability to migrate towards injured and/or inflamed tissues, and secrete an array of mediators as growth factors and cytokines, promoting tissue repair, cellular survival and differentiation. In this context, MSCs are ideal candidates for cell-based therapies, especially for the treatment of neurodegenerative diseases [155]. Although the precise mechanisms by which MSCs exert beneficial effects remain unclear, one mechanism could be the release of neurotrophic factors and other immunomodulatory compounds.

MSCs have been used in pre-clinical models of psychiatry disorders with promising results [156]. For instance, Tfilin et al. showed that treatment of an animal model of depression with MSCs increased hippocampal neurogenesis and improved depression-like behavior [157]. Intra-hippocampal transplantation of MSCs enhanced neurogenesis [158]. No clinical study using MSCs in mood disorders has been published so far. There is one clinical trial registered to use MSCs in the treatment of resistant depression (NCT02675556).

3.9 Current pharmacological treatments and inflammatory markers

Many of the current drugs approved to treat mood disorders also display some anti-inflammatory properties. For instance, antidepressants decrease peripheral levels of IL-4, IL-6, and IL-10 in MDD subjects. IL-1ß decrease was significant solely with serotonin reuptake inhibitors (SSRI), while no significant difference was observed for other cytokines such as IL-2, TNF-α, and IFN-γ. Accordingly, treatment with antidepressants may improve depressive symptoms without lowering the levels of all pro-inflammatory cytokines [159].

Lithium has also been shown to have anti-inflammatory properties. Lithium can decrease pro-inflammatory cytokines (IL-2, IL-6 and IFN-γ) while increasing anti-inflammatory cytokines as IL-4 and IL-10 in human whole-blood cell culture [160]. Chronic lithium administration to rats significantly reduced the enzyme activity of COX-2 and brain concentration of PGE2 [161].

Several studies have documented the effect of antipsychotics on inflammatory markers as well. Antipsychotic treatment leads to increased peripheral concentrations of sIL-2R and IL-12, and decreased levels of TGF-β, IL-6, and IL-1β. In an in vitro model of inflammation, toxine 1-induced release of cytokines by human whole-blood cell culture, quetiapine was shown to reduce the release of IL-2 and TNF-α, but to increase the levels of IL- 17 [162].

Ketamine, an N-methyl-D-aspartate (NMDA) receptor antagonist, has been used as a potent short-acting antidepressant in patients with severe or refractory conditions [163]. Ketamine exerts anti-inflammatory effects in both humans and animals [164, 165]. Administration of ketamine lowered the levels of IL-1β, IL-6, TNF-α, IDO (indoleamine 2,3-dioxygenase) and the kynurenine/tryptophan ratio in the rat hippocampus [166]. These anti-inflammatory effects can be linked to antidepressant activity of ketamine mainly via a direct inhibition of inflammatory cytokines and an indirect effect on the kynurenine signaling pathway [167, 168]. Similar studies have still to be carried out in patients with inflammation-associated depression.

Electroconvulsive therapy (ECT) is an effective treatment for depression, being used in medication-resistant patients or patients suffering from severe psychotic depression. IL-6 activity was increased after a single session of ECT [169]. Fluitman et al. (2011) observed that a single session of ECT was associated with increased production of IL-6, IL-10, and TNF-α, as well as decreased production of IFN-γ in peripheral monocyte cells culture [170]. After a single session of ECT, plasma IL-1β and IL-6 concentrations increased over the following 3 hours, returning to baseline concentrations in 24 h [171]. Hestad et al. found that, although ECT increased TNF-α 1 h after the first session, repeated treatments gradually reduced TNF-α that reached levels comparable to healthy controls at the end of the study [172]. Recently, Freire et al. showed that the combination of ECT with pharmacotherapy was associated with IL-6 reduction, but IFN-γ and TNF-α increase. No significant results were found for IL-2, IL-4, IL-10 and IL-17 [173]. These results suggest that ECT has significant immunomodulatory effects in patients both in the short and long-terms.

Transcranial direct current stimulation (tDCS) is a noninvasive technique that has been experimentally tested for a number of psychiatric and neurological conditions. It is based on the application of a weak direct electric current over the scalp through two electrodes: the anode-which locally increases cortical excitability, and the cathode which has opposite effects (Brunoni [174, 175]. In an animal study, tDCS decreased hippocampal TNFα levels in animals submitted to tDCS [176]. Brunoni et al. explored the effects of tDCS on immune endpoints during an antidepressant treatment in a 6-week, double-blind, placebo-controlled trial. In this study, 73 antidepressant-free patients with unipolar depression were randomized to active/sham tDCS and sertraline/placebo. Plasma levels of IL-2, IL-4, IL-6, IL-10, IL-17, IFN-γ decreased over time, but these effects were similar across the different intervention-groups and in responders vs. non-responders [177]. These studies suggest that the antidepressant effect of tDCS is not associated with peripheral modulation of immune parameters, but may be associated with immune changes in the CNS.

Deep brain stimulation (DBS) has become a therapeutic option for chronic pain, movement disorders [178], and more recently refractory depression [179]. Preclinical studies suggest that DBS may lead to alterations in immunity, with findings of increased peripheral levels of the pro-inflammatory cytokines IL-1β, IL-6, TNF-α and IFN-γ following DBS of the ventromedial hypothalamic nucleus in rats [180].

4. Expert Commentary

Current data support that a dysfunction of the immune system, mainly its innate branch, seems to be associated with mood disorders, possibly contributing to the pathophysiology of these conditions. Also, the observed imbalance in the immune system toward a persistent low-grade inflammation can be a bidirectional link between mood disorders and inflammation-related diseases such as diabetes, rheumatoid arthritis, systemic lupus erythematosus, psoriasis and autoimmune thyroiditis. Based on these facts and the ‘exhaustion’ of monoamine-based and other traditional psychopharmacological approaches, the immune system has become a target of utmost interest for the development of novel therapeutic strategies for mood disorders.

As reviewed above, several studies using immune-based therapies found positive results, i.e. reported clinical improvement, especially involving patients with depressive syndromes. The results are exciting but must be regarded as preliminary due to the low number of studies, limited sample size, and the need of replicability. Moreover, not all the findings were concordant. While distinct strategies act on different immune or inflammatory pathways (with different neurobiological implications), part of the controversial findings may be related to the heterogeneous nature of the mood disorders. Indeed, patients with high levels of inflammatory biomarkers at baseline tended to respond better to immune-base therapies in comparison with patients with normal levels of these biomarkers.

Taking into account the biological and clinical heterogeneity of MDD and BD, it is unlikely that immune dysfunction plays a major or a significant role in all cases of mood disorders. Some propose that, in at least one third of these patients, immune or inflammatory processes may be pathologically relevant. Accordingly, in these patients, anti-inflammatory or immune-based agents may represent a realistic alternative for a more personalized approach. In other words, anti-inflammatory strategies could be effective in those patients that present high levels of inflammatory markers. To confirm this hypothesis, future clinical trials must consider the inflammatory profile of the patients at the baseline and control it for possible changes with treatment. In this regard, one challenge is the definition of a valid and economically viable panel of inflammatory biomarkers. It is worth mentioning that a single inflammatory biomarker, such as the CRP, is not able to reflect a complex and multifaceted process as the inflammatory response. There has been substantial progress in the psychiatric field in an attempt to define accurate and/or sensitive biomarkers to advance the understanding of the pathophysiology of mood disorders. However, this progress was not translated into valid biomarkers for the clinical practice. Future studies must specifically address the issue of clinically meaningful biomarkers of mood disorders, including inflammation related parameters. It also remains to be determined whether specific sets of mood symptoms (e.g. atypical vs. melancholic; somatic vs. psychological) are more sensitive to immune-based strategies than others.

In conclusion, the study of immune changes in mood disorders seems to be promising to advance the understanding and the therapeutics of mood disorders. In practical terms, it highlights the relevance of optimizing the treatment of medical comorbidities, such as diabetes, metabolic syndrome and other conditions associated with a pro-inflammatory profile in patients with mood disorders corroborating the long-standing wisdom represented by the Latin expression ‘mens sana in corpore sano’ (a healthy mind in a healthy body).

5. Five-year view

Several of the treatments mentioned in this review led to positive results, improving especially depressive symptoms. As different domains compose ‘depression’, future studies may attempt to identify which ones are more (e.g. somatic symptoms) or less (e.g. subjective feelings) sensitive to the influence of anti-inflammatory strategies. Future longitudinal studies should combine different designs (alone and add-on strategies), populations (young and elderly subjects), measures (psychopathological, cognitive, functional), and surrogate markers (neuroimaging, inflammatory markers). This multimodal approach can ultimately foster the understanding of the biological basis of mood disorders, and how peripheral and immune mechanisms interact. In addition, it will help the development of more effective therapies for patients with mood disorders.

Table 1.

Immune changes observed in MDD and BD.

Major depressive disorder Bipolar disorder
Cytokines in blood ↑IL-1, ↑IL-6, ↑TNF-α, ↑CRP
↑INF-γ, ↑IL-4, ↑IL-13,
↑TNF-α, ↑INF-γ, ↑ IL-6, ↑IL-4, ↑IL-18, ↑IL-13, ↑IL-33 ↓IL-2 and ↑IL-1b, ↑CRP
Cytokines in CSF ↑IL-1β, ↓IL-6 ↑IL-1b, ↓ IL-6, ↑ IL-1β, ↑ IL-8 ↑YKL-40
Immune cells ↑mRNA IL-1β, ↑mRNA IL-6, ↑mRNA TNF-α, ↑mRNA TNFR1, ↑mRNA TNFR2,↑mRNA IL-1R1, ↑ mRNA IL-1RA ↑mRNA IL-1β, ↑mRNA IL-6, ↑mRNA TNFα, ↑mRNA IL-1R1, ↑mRNA IL-1ra, ↑mRNA TNFR1
PET studies ↑TSPO VT ↑(11)C]-(R)-PK11195
Neuropathology ↓TNFR2 mRNA, IL-1α, ↑IL-2, ↑IL-3, ↑IL-5, ↑IL-8,↑ IL-9,↑ IL-10, ↑IL-12A, ↑IL-13, ↑IL-15, ↑IL-18, ↑ IFNγ, ↑lymphotoxin α ↑mRNA IL-1β, ↑ mRNA IL-1R, ↓TNFR2 mRNA, ↑ tmTNFα, ↑GFAP,↑ iNOS, ↑c-fos, ↑CD11b
Autoimmunity disease Type 1 diabetes, multiple sclerosis, Crohn's disease, psoriasis, systemic lupus erythematosus. Autoimmune hepatitis, multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, psoriasis and autoimmune thyroiditis, Guillain-Barré syndrome.

Table 2.

Clinical trials performed for testing anti-inflammatory- / immunomodulatory-based therapies in depression.

Trial Intervention Design Outcomes Status
NCT02362529 Celecoxib and Minocycline Randomized, controlled, phase 3(n=240) No published results Currently recruiting
NCT03152409 Aspirin Randomized, controlled, phase 2 (n=74) No published results Not recruiting yet
NCT02456948 Minocycline Randomized, controlled, phase 2 (n=160) No published results Currently recruiting
NCT02263872 Minocycline Pilot Randomized Phase 4 (n=41) No published results Completed
NCT00463580 Infliximab Randomized, controlled, phase 4 (n=60) No difference in HAM-D scores between treatment groups across time However, baseline concentrations of TNF and its soluble receptors were significantly higher in infliximab-treated responders vs non responders Completed
NCT00361374 Omega-3 Fatty Acids Double-blind Randomized Placebo-controlled Phase 3 (n=196) Neither EPA-enriched nor DHA-enriched n-3 was superior to placebo for the treatment of MDD. Completed
NCT03072823 n-3 Polyunsaturated fatty acid Double-blind Randomized Placebo-controlled (n=60) No published results Currently recruiting
NCT00511810 Low Dose Fish Oil
High Dose Fish Oil
Open-Label Randomized, Phase 2,3 (n=20) LCn-3 fatty acid status of adolescent MDD patients is associated with subtle changes in glutamate + glutamine, myo-inositol, and choline concentrations in the DLPFC. Completed
NCT02553915 Omega-3 Fatty Acids Double-Blind, Randomized, Placebo-Controlled Phase 2,3 (n=100) No published results Currently recruiting
NCT00816322 Omega-3 Fatty Acids Double-Blind, Randomized, Placebo-Controlled (n=120) Patients in the omega-3 PUFA group had a significantly decreased score on the Hamilton Rating Scale for Depression than those in the placebo group. Completed
NCT00256412 Omega-3 Fatty Acids Double-Blind, Randomized, Placebo-Controlled (n=24) Omega-3 fatty acid supplementation, improve escitalopram antidepressant response. Completed
NCT00962598 Omega-3 Fatty Acids Randomized, Placebo-Controlled Phase 2 (n=57) No difference in depression severity after treatment between groups. Completed
NCT00067301 Omega-3 Fatty Acids Double-Blind, Randomized, Phase 3 (n=60) The combination therapy demonstrated significantly greater improvement in depressive symptoms. Completed
NCT00096798 Omega-3 Fatty Acids Double-Blind, Randomized, Placebo-Controlled Phase 3 (n=80) EPA demonstrated an advantage over placebo, decrease in HDRS-17 score compare to placebo. Completed
NCT01341925 Omega-3 Fatty Acids Double-Blind, Randomized, Placebo-Controlled Phase 1 (n=73) The combination of psychotherapy and Omega-3 improve behavioral problems in youth with depression Completed
NCT02972398 N-acetylcysteine Randomized, Placebo Controlled Trial (n=200) No published results Currently recruiting
NCT02469545 Probiotic Double-Blind, Randomized, (n=60) No published results Completed
NCT02838043 Probiotic Open Label Trial Phase 3 (n=10) No published results Currently recruiting
NCT02957591 Probiotic Double-Blind, Randomized, Placebo-Controlled (n=60) No published results Not recruiting yet

Table 3.

Clinical trials performed for testing anti-inflammatory- / immunomodulatory-based therapies in Bipolar Disorder.

Trial Intervention Design Outcomes Status
NCT02703363 Celecoxib and Minocycline Randomized, controlled, phase 3(n=240) No published results Currently recruiting
NCT01479829 Celecoxib Randomized, controlled, phase 4 (n=80) No published results Not recruiting yet
NCT01429272 Aspirin and Minocycline Randomized, controlled, phase 3 (n=100) No published results Completed
NCT01797575 Aspirin and NAC Double-blind Randomized Placebo-controlled (n=38) No published results Completed
NCT01403662 Minocycline Open-label Phase 3 (n=29) Adjunctive minocycline decreased depressive symptom severity from baseline to week 8. Levels IL-12/23p40 were increased, while levels of IL-12p70 and CCL26) were reduced from baseline to week 8. Completed
NCT01514422 Minocycline Open Label Phase 4 (n=20) No published results Completed
NCT02765100 Minocycline Open Label Pilot Study (n=180) No published results Currently recruiting
NCT02719392 Minocycline and NAC Double-blind Randomized Placebo-controlled Phase 4 (n=40) No published results Currently recruiting
NCT02363738 Infliximab Double-Blind, Randomized, Placebo-Controlled Phase 2 (n=60) No published results Currently recruiting
NCT00010868 Omega-3 Fatty Acids Double-Blind, Randomized, Placebo-Controlled Phase 2 (n=120) Omega 3 treatment was not superior to placebo for BD. Completed
NCT00252486 Omega-3 Fatty Acids Double-Blind, Randomized, Placebo-Controlled (n=65) There were no significant differences in primary outcome measures when compared by treatment assignment. Completed
NCT00854737 Omega-3 Fatty Acids Double-Blind, Randomized, Placebo-Controlled (n=90) There was no difference among the groups in the primary outcome Completed
NCT00001146 Omega-3 Fatty Acids Double-Blind, Randomized, Placebo-Controlled Phase 2 (n=240) There were no significant differences on any outcome measure between the EPA and placebo groups. Completed
NCT00592683 Omega-3 Fatty Acids Double-Blind, Randomized, Placebo-Controlled Phase 4 (n=20) No published results Completed
NCT02294591 N-acetylcysteine Double-Blind, Randomized, Placebo-Controlled Phase 2 (n=80) No published results Completed
NCT02719392 N-acetylcysteine Double-Blind, Randomized, Placebo-Controlled Phase 4 (n=40) No published results Currently recruiting
NCT02865629 N-acetylcysteine Open Label (n=22) No published results Currently recruiting
NCT02155972 Bifidobacterium infantis Randomized Controlled Trial Phase 2 (n=50) No published results Currently recruiting
NCT01731171 Probiotic Supplement Double-Blind, Randomized, Placebo-Controlled (n=66) No published results Completed

Key issues.

  • Over the past decade, studies have investigated the immune and/or inflammatory status of patients with MDD and BD. Compelling evidence suggests the role of inflammation in the pathophysiology of mood disorders.

  • In the context of biological and clinical heterogeneity of MDD and BD, immune dysfunction seems to plays significant role in approximately one third of the patients.

  • Several anti-inflammatory therapies have been evaluated in patients with mood disorders as adjuvant therapy.

  • ASA, minocycline and cytokine-inhibitors showed promising, but still preliminary, results in the treatment of MDD and BD.

  • Adjuvant therapies as polyunsaturated fatty acids, N-acetylcysteine, physical activity and probiotics have also shown promising effects in patients with mood disorders.

Acknowledgments

Funding

This paper was not funded.

Footnotes

Declaration of interest

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

References

Papers of special note have been highlighted as:

* of interest

** of considerable interest

  • 1.Kessler RC, Berglund P, Demler O, et al. The epidemiology of major depressive disorder: results from the National Comorbidity Survey Replication (NCS-R) JAMA. 2003 Jun 18;289(23):3095–105. doi: 10.1001/jama.289.23.3095. [DOI] [PubMed] [Google Scholar]
  • 2*.Kessler RC, Merikangas KR, Wang PS. Prevalence, comorbidity, and service utilization for mood disorders in the United States at the beginning of the twenty-first century. Annu Rev Clin Psychol. 2007;3:137–58. doi: 10.1146/annurev.clinpsy.3.022806.091444. This review highlights the high prevalence of mood disorders in the United States. [DOI] [PubMed] [Google Scholar]
  • 3.Merikangas KR, Jin R, He JP, et al. Prevalence and correlates of bipolar spectrum disorder in the world mental health survey initiative. Arch Gen Psychiatry. 2011 Mar;68(3):241–51. doi: 10.1001/archgenpsychiatry.2011.12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Kessler RC, Akiskal HS, Ames M, et al. Prevalence and effects of mood disorders on work performance in a nationally representative sample of U.S. workers. Am J Psychiatry. 2006 Sep;163(9):1561–8. doi: 10.1176/ajp.2006.163.9.1561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.WHO. World Health Organization. 2012 [Google Scholar]
  • 6.Ferrari AJ, Norman RE, Freedman G, et al. The burden attributable to mental and substance use disorders as risk factors for suicide: findings from the Global Burden of Disease Study 2010. PloS One. 2014;9(4):e91936. doi: 10.1371/journal.pone.0091936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Bolton JM, Gunnell D, Turecki G. Suicide risk assessment and intervention in people with mental illness. BMJ. 2015 Nov 09;351:h4978. doi: 10.1136/bmj.h4978. [DOI] [PubMed] [Google Scholar]
  • 8*.Goldstein BI, Carnethon MR, Matthews KA, et al. Major Depressive Disorder and Bipolar Disorder Predispose Youth to Accelerated Atherosclerosis and Early Cardiovascular Disease: A Scientific Statement From the American Heart Association. Circulation. 2015 Sep 08;132(10):965–86. doi: 10.1161/cir.0000000000000229. This is a statement paper from the AHA about cardiovascular comorbidities in mood disorders. [DOI] [PubMed] [Google Scholar]
  • 9.Rush AJ, Trivedi MH, Wisniewski SR, et al. Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report. Am J Psychiatry. 2006 Nov;163(11):1905–17. doi: 10.1176/ajp.2006.163.11.1905. [DOI] [PubMed] [Google Scholar]
  • 10.Souery D, Papakostas GI, Trivedi MH. Treatment-resistant depression. J Clin Psychiatry. 2006;67(Suppl 6):16–22. [PubMed] [Google Scholar]
  • 11.Murrough JW, Wade E, Sayed S, et al. Dextromethorphan/quinidine pharmacotherapy in patients with treatment resistant depression: A proof of concept clinical trial. J Affect Disord. 2017 Aug 15;218:277–283. doi: 10.1016/j.jad.2017.04.072. [DOI] [PubMed] [Google Scholar]
  • 12.Pangalos MN, Schechter LE, Hurko O. Drug development for CNS disorders: strategies for balancing risk and reducing attrition. Nat Rev Drug Discov. 2007 Jul;6(7):521–32. doi: 10.1038/nrd2094. [DOI] [PubMed] [Google Scholar]
  • 13.Berton O, Nestler EJ. New approaches to antidepressant drug discovery: beyond monoamines. Nat Rev Neurosci. 2006 Feb;7(2):137–51. doi: 10.1038/nrn1846. [DOI] [PubMed] [Google Scholar]
  • 14*.Papakostas GI, Ionescu DF. Towards new mechanisms: an update on therapeutics for treatment-resistant major depressive disorder. Mol Psychiatry. 2015 Oct;20(10):1142–50. doi: 10.1038/mp.2015.92. This is a recent review on new antidepressant approaches. [DOI] [PubMed] [Google Scholar]
  • 15.Caban A, Pisarczyk K, Kopacz K, et al. Filling the gap in CNS drug development: evaluation of the role of drug repurposing. J Mark Access Health Policy. 2017;5(1):1299833. doi: 10.1080/20016689.2017.1299833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16*.Teixeira AL, Muller N. Immunology of psychiatric disorders. Neuroimmunomodulation. 2014;21(2–3):71. doi: 10.1159/000356525. This supplement reviews the immunological changes observed in patients with major psychiatric disorders. [DOI] [PubMed] [Google Scholar]
  • 17.Noto C, Rizzo LB, Mansur RB, et al. Targeting the inflammatory pathway as a therapeutic tool for major depression. Neuroimmunomodulation. 2014;21(2–3):131–9. doi: 10.1159/000356549. [DOI] [PubMed] [Google Scholar]
  • 18.Haapakoski R, Ebmeier KP, Alenius H, et al. Innate and adaptive immunity in the development of depression: An update on current knowledge and technological advances. Prog Neuropsychopharmacol Biol Psychiatry. 2016 Apr 03;66:63–72. doi: 10.1016/j.pnpbp.2015.11.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Sayana P, Colpo GD, Simoes LR, et al. A systematic review of evidence for the role of inflammatory biomarkers in bipolar patients. J Psychiatr Res. 2017 Sep;92:160–182. doi: 10.1016/j.jpsychires.2017.03.018. [DOI] [PubMed] [Google Scholar]
  • 20.Schmidt FM, Lichtblau N, Minkwitz J, et al. Cytokine levels in depressed and non-depressed subjects, and masking effects of obesity. J Psychiatr Res. 2014 Aug;55:29–34. doi: 10.1016/j.jpsychires.2014.04.021. [DOI] [PubMed] [Google Scholar]
  • 21**.Dahl J, Ormstad H, Aass HC, et al. The plasma levels of various cytokines are increased during ongoing depression and are reduced to normal levels after recovery. Psychoneuroendocrinology. 2014 Jul;45:77–86. doi: 10.1016/j.psyneuen.2014.03.019. This study shows the decrease of inflammatory markers with the treatment of depression. [DOI] [PubMed] [Google Scholar]
  • 22.Howren MB, Lamkin DM, Suls J. Associations of depression with C-reactive protein, IL-1, and IL-6: a meta-analysis. Psychosomatic medicine. 2009 Feb;71(2):171–86. doi: 10.1097/PSY.0b013e3181907c1b. [DOI] [PubMed] [Google Scholar]
  • 23**.Liu Y, Ho RC, Mak A. Interleukin (IL)-6, tumour necrosis factor alpha (TNF-alpha) and soluble interleukin-2 receptors (sIL-2R) are elevated in patients with major depressive disorder: a meta-analysis and meta-regression. J Affect Disord. 2012 Aug;139(3):230–9. doi: 10.1016/j.jad.2011.08.003. This meta-analysis corroborates that inflammatory markers are increased in MDD. [DOI] [PubMed] [Google Scholar]
  • 24.Dowlati Y, Herrmann N, Swardfager W, et al. A meta-analysis of cytokines in major depression. Biol Psychiatry. 2010 Mar 01;67(5):446–57. doi: 10.1016/j.biopsych.2009.09.033. [DOI] [PubMed] [Google Scholar]
  • 25.Courtet P, Jaussent I, Genty C, et al. Increased CRP levels may be a trait marker of suicidal attempt. European Neuropsychopharmacology. 2015 Oct;25(10):1824–31. doi: 10.1016/j.euroneuro.2015.05.003. [DOI] [PubMed] [Google Scholar]
  • 26.O’Donovan A, Rush G, Hoatam G, et al. Suicidal ideation is associated with elevated inflammation in patients with major depressive disorder. Depress anxiety. 2013 Apr;30(4):307–14. doi: 10.1002/da.22087. [DOI] [PubMed] [Google Scholar]
  • 27.Levine J, Barak Y, Chengappa KN, et al. Cerebrospinal cytokine levels in patients with acute depression. Neuropsychobiology. 1999 Nov;40(4):171–6. doi: 10.1159/000026615. doi: 26615. [DOI] [PubMed] [Google Scholar]
  • 28.Stubner S, Schon T, Padberg F, et al. Interleukin-6 and the soluble IL-6 receptor are decreased in cerebrospinal fluid of geriatric patients with major depression: no alteration of soluble gp130. Neuroscience letters. 1999 Jan 15;259(3):145–8. doi: 10.1016/s0304-3940(98)00916-1. [DOI] [PubMed] [Google Scholar]
  • 29.Carpenter LL, Heninger GR, Malison RT, et al. Cerebrospinal fluid interleukin (IL)-6 in unipolar major depression. J Affect Disord. 2004 Apr;79(1–3):285–9. doi: 10.1016/s0165-0327(02)00460-3. [DOI] [PubMed] [Google Scholar]
  • 30.Dean B, Gibbons AS, Tawadros N, et al. Different changes in cortical tumor necrosis factor-alpha-related pathways in schizophrenia and mood disorders. Mol Psychiatry. 2013 Jul;18(7):767–73. doi: 10.1038/mp.2012.95. [DOI] [PubMed] [Google Scholar]
  • 31.Kim S, Hwang Y, Webster MJ, et al. Differential activation of immune/inflammatory response-related co-expression modules in the hippocampus across the major psychiatric disorders. Mol Psychiatry. 2016 Mar;21(3):376–85. doi: 10.1038/mp.2015.79. [DOI] [PubMed] [Google Scholar]
  • 32.Torres-Platas SG, Cruceanu C, Chen GG, et al. Evidence for increased microglial priming and macrophage recruitment in the dorsal anterior cingulate white matter of depressed suicides. Brain Behav Immun. 2014 Nov;42:50–9. doi: 10.1016/j.bbi.2014.05.007. [DOI] [PubMed] [Google Scholar]
  • 33.Setiawan E, Wilson AA, Mizrahi R, et al. Role of translocator protein density, a marker of neuroinflammation, in the brain during major depressive episodes. JAMA psychiatry. 2015 Mar;72(3):268–75. doi: 10.1001/jamapsychiatry.2014.2427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Walsh JG, Muruve DA, Power C. Inflammasomes in the CNS. Nature reviews Neuroscience. 2014 Feb;15(2):84–97. doi: 10.1038/nrn3638. [DOI] [PubMed] [Google Scholar]
  • 35.Sousa C, Biber K, Michelucci A. Cellular and Molecular Characterization of Microglia: A Unique Immune Cell Population. Front Immunol. 2017;8:198. doi: 10.3389/fimmu.2017.00198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Bhattacharya A, Derecki NC, Lovenberg TW, et al. Role of neuro-immunological factors in the pathophysiology of mood disorders. Psychopharmacology. 2016 May;233(9):1623–36. doi: 10.1007/s00213-016-4214-0. [DOI] [PubMed] [Google Scholar]
  • 37.Bitzer-Quintero OK, Gonzalez-Burgos I. Immune system in the brain: a modulatory role on dendritic spine morphophysiology? Neural Plast. 2012;2012:348642. doi: 10.1155/2012/348642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Patel JP, Frey BN. Disruption in the Blood-Brain Barrier: The Missing Link between Brain and Body Inflammation in Bipolar Disorder? Neural Plast. 2015;2015:708306. doi: 10.1155/2015/708306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Dantzer R, Konsman JP, Bluthe RM, et al. Neural and humoral pathways of communication from the immune system to the brain: parallel or convergent? Auton Neurosci. 2000 Dec 20;85(1–3):60–5. doi: 10.1016/s1566-0702(00)00220-4. [DOI] [PubMed] [Google Scholar]
  • 40.D’Mello C, Le T, Swain MG. Cerebral microglia recruit monocytes into the brain in response to tumor necrosis factoralpha signaling during peripheral organ inflammation. J Neurosci. 2009 Feb 18;29(7):2089–102. doi: 10.1523/jneurosci.3567-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Menard C, Hodes GE, Russo SJ. Pathogenesis of depression: Insights from human and rodent studies. Neuroscience. 2016 May 03;321:138–62. doi: 10.1016/j.neuroscience.2015.05.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Korczak DJ, Pereira S, Koulajian K, et al. Type 1 diabetes mellitus and major depressive disorder: evidence for a biological link. Diabetologia. 2011 Oct;54(10):2483–93. doi: 10.1007/s00125-011-2240-3. [DOI] [PubMed] [Google Scholar]
  • 43.Vonk R, van der Schot AC, Kahn RS, et al. Is autoimmune thyroiditis part of the genetic vulnerability (or an endophenotype) for bipolar disorder? Biol Psychiatry. 2007 Jul 15;62(2):135–40. doi: 10.1016/j.biopsych.2006.08.041. [DOI] [PubMed] [Google Scholar]
  • 44.Wulsin LR, Singal BM. Do depressive symptoms increase the risk for the onset of coronary disease? A systematic quantitative review. Psychosomatic medicine. 2003 Mar-Apr;65(2):201–10. doi: 10.1097/01.psy.0000058371.50240.e3. [DOI] [PubMed] [Google Scholar]
  • 45.Van der Kooy K, van Hout H, Marwijk H, et al. Depression and the risk for cardiovascular diseases: systematic review and meta analysis. Int J Ger Psychiatry. 2007 Jul;22(7):613–26. doi: 10.1002/gps.1723. [DOI] [PubMed] [Google Scholar]
  • 46.Rugulies R. Depression as a predictor for coronary heart disease. a review and meta-analysis. Am J Prev Med. 2002 Jul;23(1):51–61. doi: 10.1016/s0749-3797(02)00439-7. [DOI] [PubMed] [Google Scholar]
  • 47.Valkanova V, Ebmeier KP. Vascular risk factors and depression in later life: a systematic review and meta-analysis. Biol Psychiatry. 2013 Mar 01;73(5):406–13. doi: 10.1016/j.biopsych.2012.10.028. [DOI] [PubMed] [Google Scholar]
  • 48.Kunugi H, Hori H, Ogawa S. Biochemical markers subtyping major depressive disorder. Psychiatry Clin Neurosci. 2015 Oct;69(10):597–608. doi: 10.1111/pcn.12299. [DOI] [PubMed] [Google Scholar]
  • 49.Gold PW. The organization of the stress system and its dysregulation in depressive illness. Mol Psychiatry. 2015 Feb;20(1):32–47. doi: 10.1038/mp.2014.163. [DOI] [PubMed] [Google Scholar]
  • 50.Lamers F, Vogelzangs N, Merikangas KR, et al. Evidence for a differential role of HPA-axis function, inflammation and metabolic syndrome in melancholic versus atypical depression. Mol Psychiatry. 2013 Jun;18(6):692–9. doi: 10.1038/mp.2012.144. [DOI] [PubMed] [Google Scholar]
  • 51.Cho HJ, Seeman TE, Bower JE, et al. Prospective association between C-reactive protein and fatigue in the coronary artery risk development in young adults study. Biol Psychiatry. 2009 Nov 01;66(9):871–8. doi: 10.1016/j.biopsych.2009.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Capuron L, Su S, Miller AH, et al. Depressive symptoms and metabolic syndrome: is inflammation the underlying link? Biol Psychiatry. 2008 Nov 15;64(10):896–900. doi: 10.1016/j.biopsych.2008.05.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Soderlund J, Olsson SK, Samuelsson M, et al. Elevation of cerebrospinal fluid interleukin-1ss in bipolar disorder. J Psychiatry Neurosci : JPN. 2011 Mar;36(2):114–8. doi: 10.1503/jpn.100080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Rolstad S, Jakobsson J, Sellgren C, et al. CSF neuroinflammatory biomarkers in bipolar disorder are associated with cognitive impairment. European neuropsychopharmacology. 2015 Aug;25(8):1091–8. doi: 10.1016/j.euroneuro.2015.04.023. [DOI] [PubMed] [Google Scholar]
  • 55*.Jakobsson J, Bjerke M, Sahebi S, et al. Monocyte and microglial activation in patients with mood-stabilized bipolar disorder. J Psychiatry Neurosci : JPN. 2015 Jul;40(4):250–8. doi: 10.1503/jpn.140183. This paper shows evidence of microglial activation in patients with bipolar disorder. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Brietzke E, Kauer-Sant’Anna M, Teixeira AL, et al. Abnormalities in serum chemokine levels in euthymic patients with bipolar disorder. Brain Behav Immun. 2009 Nov;23(8):1079–82. doi: 10.1016/j.bbi.2009.04.008. [DOI] [PubMed] [Google Scholar]
  • 57.Brietzke E, Stertz L, Fernandes BS, et al. Comparison of cytokine levels in depressed, manic and euthymic patients with bipolar disorder. J Affect Disord. 2009 Aug;116(3):214–7. doi: 10.1016/j.jad.2008.12.001. [DOI] [PubMed] [Google Scholar]
  • 58.Maes M, Bosmans E, Calabrese J, et al. Interleukin-2 and interleukin-6 in schizophrenia and mania: effects of neuroleptics and mood stabilizers. J Psychiatr Res. 1995 Mar-Apr;29(2):141–52. doi: 10.1016/0022-3956(94)00049-w. [DOI] [PubMed] [Google Scholar]
  • 59.Kim YK, Jung HG, Myint AM, et al. Imbalance between pro-inflammatory and anti-inflammatory cytokines in bipolar disorder. J Affect Disord. 2007 Dec;104(1–3):91–5. doi: 10.1016/j.jad.2007.02.018. [DOI] [PubMed] [Google Scholar]
  • 60.O’Brien SM, Scully P, Scott LV, et al. Cytokine profiles in bipolar affective disorder: focus on acutely ill patients. J Affect Disord. 2006 Feb;90(2–3):263–7. doi: 10.1016/j.jad.2005.11.015. [DOI] [PubMed] [Google Scholar]
  • 61.Ortiz-Dominguez A, Hernandez ME, Berlanga C, et al. Immune variations in bipolar disorder: phasic differences. Bipolar Disord. 2007 Sep;9(6):596–602. doi: 10.1111/j.1399-5618.2007.00493.x. [DOI] [PubMed] [Google Scholar]
  • 62.Padmos RC, Hillegers MH, Knijff EM, et al. A discriminating messenger RNA signature for bipolar disorder formed by an aberrant expression of inflammatory genes in monocytes. Arch Gen Psychiatry. 2008 Apr;65(4):395–407. doi: 10.1001/archpsyc.65.4.395. [DOI] [PubMed] [Google Scholar]
  • 63.Breunis MN, Kupka RW, Nolen WA, et al. High numbers of circulating activated T cells and raised levels of serum IL-2 receptor in bipolar disorder. Biol Psychiatry. 2003 Jan 15;53(2):157–65. doi: 10.1016/s0006-3223(02)01452-x. [DOI] [PubMed] [Google Scholar]
  • 64.Tsai SY, Yang YY, Kuo CJ, et al. Effects of symptomatic severity on elevation of plasma soluble interleukin-2 receptor in bipolar mania. J Affect Disord. 2001 May;64(2–3):185–93. doi: 10.1016/s0165-0327(00)00252-4. [DOI] [PubMed] [Google Scholar]
  • 65.Tsai SY, Chen KP, Yang YY, et al. Activation of indices of cell-mediated immunity in bipolar mania. Biol Psychiatry. 1999 Apr 15;45(8):989–94. doi: 10.1016/s0006-3223(98)00159-0. [DOI] [PubMed] [Google Scholar]
  • 66.Cunha AB, Andreazza AC, Gomes FA, et al. Investigation of serum high-sensitive C-reactive protein levels across all mood states in bipolar disorder. Eur Arch Psychiatry Clin Neurosci. 2008 Aug;258(5):300–4. doi: 10.1007/s00406-007-0797-0. [DOI] [PubMed] [Google Scholar]
  • 67.Huang TL, Lin FC. High-sensitivity C-reactive protein levels in patients with major depressive disorder and bipolar mania. Prog Neuropsychopharmacol Biol Psychiatry. 2007 Mar 30;31(2):370–2. doi: 10.1016/j.pnpbp.2006.09.010. [DOI] [PubMed] [Google Scholar]
  • 68.Wadee AA, Kuschke RH, Wood LA, et al. Serological observations in patients suffering from acute manic episodes. Hum Psychopharmacol. 2002 Jun;17(4):175–9. doi: 10.1002/hup.390. [DOI] [PubMed] [Google Scholar]
  • 69.Tsai SY, Chung KH, Wu JY, et al. Inflammatory markers and their relationships with leptin and insulin from acute mania to full remission in bipolar disorder. J Affect Disord. 2012 Jan;136(1–2):110–116. doi: 10.1016/j.jad.2011.08.022. [DOI] [PubMed] [Google Scholar]
  • 70*.Rao JS, Harry GJ, Rapoport SI, et al. Increased excitotoxicity and neuroinflammatory markers in postmortem frontal cortex from bipolar disorder patients. Mol Psychiatry. 2010 Apr;15(4):384–92. doi: 10.1038/mp.2009.47. This paper reported neuroinflammation in brain from BD patients. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Haarman BC, Riemersma-Van der Lek RF, de Groot JC, et al. Neuroinflammation in bipolar disorder - A [(11)C]-(R)-PK11195 positron emission tomography study. Brain Behav Immun. 2014 Aug;40:219–25. doi: 10.1016/j.bbi.2014.03.016. [DOI] [PubMed] [Google Scholar]
  • 72.Haarman BC, Burger H, Doorduin J, et al. Volume, metabolites and neuroinflammation of the hippocampus in bipolar disorder - A combined magnetic resonance imaging and positron emission tomography study. Brain Behav Immun. 2016 Aug;56:21–33. doi: 10.1016/j.bbi.2015.09.004. [DOI] [PubMed] [Google Scholar]
  • 73.Pandey GN, Ren X, Rizavi HS, et al. Abnormal gene expression of proinflammatory cytokines and their receptors in the lymphocytes of patients with bipolar disorder. Bipolar Disord. 2015 Sep;17(6):636–44. doi: 10.1111/bdi.12320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Barbosa IG, Rocha NP, Assis F, et al. Monocyte and lymphocyte activation in bipolar disorder: a new piece in the puzzle of immune dysfunction in mood disorders. Int J Neuropsychopharmacol. 2014 Oct 31;18(1) doi: 10.1093/ijnp/pyu021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Barbosa IG, Nogueira CR, Rocha NP, et al. Altered intracellular signaling cascades in peripheral blood mononuclear cells from BD patients. J Psychiatr Res. 2013 Dec;47(12):1949–54. doi: 10.1016/j.jpsychires.2013.08.019. [DOI] [PubMed] [Google Scholar]
  • 76.SayuriYamagata A, Brietzke E, Rosenblat JD, et al. Medical comorbidity in bipolar disorder: The link with metabolic-inflammatory systems. J Affect Disord. 2017 Mar 15;211:99–106. doi: 10.1016/j.jad.2016.12.059. [DOI] [PubMed] [Google Scholar]
  • 77.Hillegers MH, Reichart CG, Wals M, et al. Signs of a higher prevalence of autoimmune thyroiditis in female offspring of bipolar parents. European neuropsychopharmacology. 2007 May-Jun;17(6–7):394–9. doi: 10.1016/j.euroneuro.2006.10.005. [DOI] [PubMed] [Google Scholar]
  • 78.Zhao Z, Okusaga OO, Quevedo J, et al. The potential association between obesity and bipolar disorder: A meta-analysis. J Affect Disord. 2016 Sep 15;202:120–3. doi: 10.1016/j.jad.2016.05.059. [DOI] [PubMed] [Google Scholar]
  • 79.Vane JR, Botting RM. The mechanism of action of aspirin. Thromb Res. 2003 Jun 15;110(5–6):255–8. doi: 10.1016/s0049-3848(03)00379-7. [DOI] [PubMed] [Google Scholar]
  • 80*.Abbasi SH, Hosseini F, Modabbernia A, et al. Effect of celecoxib add-on treatment on symptoms and serum IL-6 concentrations in patients with major depressive disorder: randomized double-blind placebo-controlled study. J Affect Disord. 2012 Dec 10;141(2–3):308–14. doi: 10.1016/j.jad.2012.03.033. Clinical trial with celecoxib in patients with MDD. [DOI] [PubMed] [Google Scholar]
  • 81.Muller N, Schwarz MJ, 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 Jul;11(7):680–4. doi: 10.1038/sj.mp.4001805. [DOI] [PubMed] [Google Scholar]
  • 82.Akhondzadeh S, Jafari S, Raisi F, et al. Clinical trial of adjunctive celecoxib treatment in patients with major depression: a double blind and placebo controlled trial. Depress anxiety. 2009;26(7):607–11. doi: 10.1002/da.20589. [DOI] [PubMed] [Google Scholar]
  • 83.Nery FG, Monkul ES, Hatch JP, et al. Celecoxib as an adjunct in the treatment of depressive or mixed episodes of bipolar disorder: a double-blind, randomized, placebo-controlled study. Human psychopharmacology. 2008 Mar;23(2):87–94. doi: 10.1002/hup.912. [DOI] [PubMed] [Google Scholar]
  • 84.Maes M. Targeting cyclooxygenase-2 in depression is not a viable therapeutic approach and may even aggravate the pathophysiology underpinning depression. Metab Brain Dis. 2012 Dec;27(4):405–13. doi: 10.1007/s11011-012-9326-6. [DOI] [PubMed] [Google Scholar]
  • 85.Cipollone F, Patrignani P, Greco A, et al. Differential suppression of thromboxane biosynthesis by indobufen and aspirin in patients with unstable angina. Circulation. 1997 Aug 19;96(4):1109–16. doi: 10.1161/01.cir.96.4.1109. [DOI] [PubMed] [Google Scholar]
  • 86.Chandrasekharan JA, Sharma-Walia N. Lipoxins: nature’s way to resolve inflammation. J Inflamm Res. 2015;8:181–92. doi: 10.2147/jir.s90380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Mendlewicz J, Kriwin P, Oswald P, et al. Shortened onset of action of antidepressants in major depression using acetylsalicylic acid augmentation: a pilot open-label study. Int Clin Psychopharmacol. 2006 Jul;21(4):227–31. doi: 10.1097/00004850-200607000-00005. [DOI] [PubMed] [Google Scholar]
  • 88.Almeida OP, Alfonso H, Jamrozik K, et al. Aspirin use, depression, and cognitive impairment in later life: the health in men study. J Am Geriatr Soc. 2010 May;58(5):990–2. doi: 10.1111/j.1532-5415.2010.02827.x. [DOI] [PubMed] [Google Scholar]
  • 89.Stolk P, Souverein PC, Wilting I, et al. Is aspirin useful in patients on lithium? A pharmacoepidemiological study related to bipolar disorder. Prostaglandins Leukot Essent Fatty Acids. 2010 Jan;82(1):9–14. doi: 10.1016/j.plefa.2009.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Levine J, Cholestoy A, Zimmerman J. Possible antidepressant effect of minocycline. Am J Psychiatry. 1996 Apr;153(4):582. doi: 10.1176/ajp.153.4.582b. [DOI] [PubMed] [Google Scholar]
  • 91.Soczynska JK, Mansur RB, Brietzke E, et al. Novel therapeutic targets in depression: minocycline as a candidate treatment. Behav Brain Res. 2012 Dec 01;235(2):302–17. doi: 10.1016/j.bbr.2012.07.026. [DOI] [PubMed] [Google Scholar]
  • 92.Homsi S, Federico F, Croci N, et al. Minocycline effects on cerebral edema: relations with inflammatory and oxidative stress markers following traumatic brain injury in mice. Brain Res. 2009 Sep 29;1291:122–32. doi: 10.1016/j.brainres.2009.07.031. [DOI] [PubMed] [Google Scholar]
  • 93.Kim SS, Kong PJ, Kim BS, et al. Inhibitory action of minocycline on lipopolysaccharide-induced release of nitric oxide and prostaglandin E2 in BV2 microglial cells. Arch Pharm Res. 2004 Mar;27(3):314–8. doi: 10.1007/BF02980066. [DOI] [PubMed] [Google Scholar]
  • 94.Tomas-Camardiel M, Rite I, Herrera AJ, et al. Minocycline reduces the lipopolysaccharide-induced inflammatory reaction, peroxynitrite-mediated nitration of proteins, disruption of the blood-brain barrier, and damage in the nigral dopaminergic system. Neurobiol Dis. 2004 Jun;16(1):190–201. doi: 10.1016/j.nbd.2004.01.010. [DOI] [PubMed] [Google Scholar]
  • 95.Miyaoka T, Wake R, Furuya M, et al. Minocycline as adjunctive therapy for patients with unipolar psychotic depression: an open-label study. Prog Neuropsychopharmacol Biol Psychiatry. 2012 Jun 01;37(2):222–6. doi: 10.1016/j.pnpbp.2012.02.002. [DOI] [PubMed] [Google Scholar]
  • 96**.Dantzer R, O’Connor JC, Freund GG, et al. From inflammation to sickness and depression: when the immune system subjugates the brain. Nat Rev Neurosci. 2008 Jan;9(1):46–56. doi: 10.1038/nrn2297. This manuscript reviews the biological and phenomenological similarities between sickness behavior and depression. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Ertenli I, Ozer S, Kiraz S, et al. Infliximab, a TNF-alpha antagonist treatment in patients with ankylosing spondylitis: the impact on depression, anxiety and quality of life level. Rheumatol Int. 2012 Feb;32(2):323–30. doi: 10.1007/s00296-010-1616-x. [DOI] [PubMed] [Google Scholar]
  • 98.Feldman SR, Gottlieb AB, Bala M, et al. Infliximab improves health-related quality of life in the presence of comorbidities among patients with moderate-to-severe psoriasis. Br J Dermatol. 2008 Sep;159(3):704–10. doi: 10.1111/j.1365-2133.2008.08727.x. [DOI] [PubMed] [Google Scholar]
  • 99.Persoons P, Vermeire S, Demyttenaere K, et al. The impact of major depressive disorder on the short- and long-term outcome of Crohn’s disease treatment with infliximab. Aliment Pharmacol Ther. 2005 Jul 15;22(2):101–10. doi: 10.1111/j.1365-2036.2005.02535.x. [DOI] [PubMed] [Google Scholar]
  • 100*.Kappelmann N, Lewis G, Dantzer R, et al. Antidepressant activity of anti-cytokine treatment: a systematic review and meta-analysis of clinical trials of chronic inflammatory conditions. Mol Psychiatry. 2016 Oct 18; doi: 10.1038/mp.2016.167. Meta-analysis on antidepressant activity of anti-cytokine treatments. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Raison CL, Rutherford RE, Woolwine BJ, et al. A randomized controlled trial of the tumor necrosis factor antagonist infliximab for treatment-resistant depression: the role of baseline inflammatory biomarkers. JAMA Psychiatry. 2013 Jan;70(1):31–41. doi: 10.1001/2013.jamapsychiatry.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Atzeni F, Talotta R, Salaffi F, et al. Immunogenicity and autoimmunity during anti-TNF therapy. Autoimmun Rev. 2013 May;12(7):703–8. doi: 10.1016/j.autrev.2012.10.021. [DOI] [PubMed] [Google Scholar]
  • 103.Bongartz T, Sutton AJ, Sweeting MJ, et al. Anti-TNF antibody therapy in rheumatoid arthritis and the risk of serious infections and malignancies: systematic review and meta-analysis of rare harmful effects in randomized controlled trials. JAMA. 2006 May 17;295(19):2275–85. doi: 10.1001/jama.295.19.2275. [DOI] [PubMed] [Google Scholar]
  • 104.van Dartel SA, Fransen J, Kievit W, et al. Difference in the risk of serious infections in patients with rheumatoid arthritis treated with adalimumab, infliximab and etanercept: results from the Dutch Rheumatoid Arthritis Monitoring (DREAM) registry. Ann Rheum Dis. 2013 Jun;72(6):895–900. doi: 10.1136/annrheumdis-2012-201338. [DOI] [PubMed] [Google Scholar]
  • 105.Simopoulos AP. Omega-3 fatty acids in health and disease and in growth and development. Am J Clin Nutr. 1991 Sep;54(3):438–63. doi: 10.1093/ajcn/54.3.438. [DOI] [PubMed] [Google Scholar]
  • 106.Calder PC. Polyunsaturated fatty acids, inflammatory processes and inflammatory bowel diseases. Mol Nutr Food Res. 2008 Aug;52(8):885–97. doi: 10.1002/mnfr.200700289. [DOI] [PubMed] [Google Scholar]
  • 107.Kiecolt-Glaser JK, Belury MA, Andridge R, et al. Omega-3 supplementation lowers inflammation in healthy middle-aged and older adults: a randomized controlled trial. Brain Behav Immun. 2012 Aug;26(6):988–95. doi: 10.1016/j.bbi.2012.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Kiecolt-Glaser JK, Belury MA, Andridge R, et al. Omega-3 supplementation lowers inflammation and anxiety in medical students: a randomized controlled trial. Brain Behav Immun. 2011 Nov;25(8):1725–34. doi: 10.1016/j.bbi.2011.07.229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Appleton KM, Rogers PJ, Ness AR. Updated systematic review and meta-analysis of the effects of n-3 long-chain polyunsaturated fatty acids on depressed mood. Am J Clin Nutr. 2010 Mar;91(3):757–70. doi: 10.3945/ajcn.2009.28313. [DOI] [PubMed] [Google Scholar]
  • 110.Martins JG, Bentsen H, Puri BK. Eicosapentaenoic acid appears to be the key omega-3 fatty acid component associated with efficacy in major depressive disorder: a critique of Bloch and Hannestad and updated meta-analysis. Mol Psychiatry. 2012 Dec;17(12):1144–9. doi: 10.1038/mp.2012.25. discussion 1163–7. [DOI] [PubMed] [Google Scholar]
  • 111.Gertsik L, Poland RE, Bresee C, et al. Omega-3 fatty acid augmentation of citalopram treatment for patients with major depressive disorder. J Clin Psychopharmacol. 2012 Feb;32(1):61–4. doi: 10.1097/JCP.0b013e31823f3b5f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Jazayeri S, Tehrani-Doost M, Keshavarz SA, et al. Comparison of therapeutic effects of omega-3 fatty acid eicosapentaenoic acid and fluoxetine, separately and in combination, in major depressive disorder. Aust N Z J Psychiatry. 2008 Mar;42(3):192–8. doi: 10.1080/00048670701827275. [DOI] [PubMed] [Google Scholar]
  • 113.Peet M, Horrobin DF. A dose-ranging study of the effects of ethyl-eicosapentaenoate in patients with ongoing depression despite apparently adequate treatment with standard drugs. Arch Gen Psychiatry. 2002 Oct;59(10):913–9. doi: 10.1001/archpsyc.59.10.913. [DOI] [PubMed] [Google Scholar]
  • 114.Stoll AL, Severus WE, Freeman MP, et al. Omega 3 fatty acids in bipolar disorder: a preliminary double-blind, placebo-controlled trial. Arch Gen Psychiatry. 1999 May;56(5):407–12. doi: 10.1001/archpsyc.56.5.407. [DOI] [PubMed] [Google Scholar]
  • 115.Hirashima F, Parow AM, Stoll AL, et al. Omega-3 fatty acid treatment and T(2) whole brain relaxation times in bipolar disorder. Am J Psychiatry. 2004 Oct;161(10):1922–4. doi: 10.1176/ajp.161.10.1922. [DOI] [PubMed] [Google Scholar]
  • 116.Keck PE, Jr, Mintz J, McElroy SL, et al. Double-blind, randomized, placebo-controlled trials of ethyl-eicosapentanoate in the treatment of bipolar depression and rapid cycling bipolar disorder. Biol Psychiatry. 2006 Nov 01;60(9):1020–2. doi: 10.1016/j.biopsych.2006.03.056. [DOI] [PubMed] [Google Scholar]
  • 117.Frangou S, Lewis M, McCrone P. Efficacy of ethyl-eicosapentaenoic acid in bipolar depression: randomised double-blind placebo-controlled study. Br J Psychiatry. 2006 Jan;188:46–50. doi: 10.1192/bjp.188.1.46. [DOI] [PubMed] [Google Scholar]
  • 118.Frangou S, Lewis M, Wollard J, et al. Preliminary in vivo evidence of increased N-acetyl-aspartate following eicosapentanoic acid treatment in patients with bipolar disorder. J Psychopharmacol (Oxford, England) 2007 Jun;21(4):435–9. doi: 10.1177/0269881106067787. [DOI] [PubMed] [Google Scholar]
  • 119.Chiu CC, Huang SY, Chen CC, et al. Omega-3 fatty acids are more beneficial in the depressive phase than in the manic phase in patients with bipolar I disorder. J Clin Psychiatry. 2005 Dec;66(12):1613–4. doi: 10.4088/jcp.v66n1219b. [DOI] [PubMed] [Google Scholar]
  • 120.Jacka FN, O’Neil A, Opie R, et al. A randomised controlled trial of dietary improvement for adults with major depression (the ‘SMILES’ trial) BMC Med. 2017 Jan 30;15(1):23. doi: 10.1186/s12916-017-0791-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Liu X, Yan Y, Li F, et al. Fruit and vegetable consumption and the risk of depression: A meta-analysis. Nutrition (Burbank, Los Angeles County, Calif) 2016 Mar;32(3):296–302. doi: 10.1016/j.nut.2015.09.009. [DOI] [PubMed] [Google Scholar]
  • 122.Rajkowska G, Miguel-Hidalgo JJ. Gliogenesis and glial pathology in depression. CNS Neurol Disord Drug Targets s. 2007 Jun;6(3):219–33. doi: 10.2174/187152707780619326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Tsai GY, Cui JZ, Syed H, et al. Effect of N-acetylcysteine on the early expression of inflammatory markers in the retina and plasma of diabetic rats. Clin Exp Ophthalmol. 2009 Mar;37(2):223–31. doi: 10.1111/j.1442-9071.2009.02000.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Kigerl KA, Ankeny DP, Garg SK, et al. System x(c)(-) regulates microglia and macrophage glutamate excitotoxicity in vivo. Exp Neurol. 2012 Jan;233(1):333–41. doi: 10.1016/j.expneurol.2011.10.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Berk M, Dean OM, Cotton SM, et al. The efficacy of adjunctive N-acetylcysteine in major depressive disorder: a double-blind, randomized, placebo-controlled trial. J Clin Psychiatry. 2014 Jun;75(6):628–36. doi: 10.4088/JCP.13m08454. [DOI] [PubMed] [Google Scholar]
  • 126*.Berk M, Copolov DL, Dean O, et al. N-acetyl cysteine for depressive symptoms in bipolar disorder–a double-blind randomized placebo-controlled trial. Biol Psychiatry. 2008 Sep 15;64(6):468–75. doi: 10.1016/j.biopsych.2008.04.022. Clinical trial with N-acetylcysteine in BD patients with depressive symptoms. [DOI] [PubMed] [Google Scholar]
  • 127.Berk M, Dean O, Cotton SM, et al. The efficacy of N-acetylcysteine as an adjunctive treatment in bipolar depression: an open label trial. J Affect Disord. 2011 Dec;135(1–3):389–94. doi: 10.1016/j.jad.2011.06.005. [DOI] [PubMed] [Google Scholar]
  • 128.Mora S, Cook N, Buring JE, et al. Physical activity and reduced risk of cardiovascular events: potential mediating mechanisms. Circulation. 2007 Nov 06;116(19):2110–8. doi: 10.1161/CIRCULATIONAHA.107.729939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Schuch FB, Vancampfort D, Rosenbaum S, et al. Exercise for depression in older adults: a meta-analysis of randomized controlled trials adjusting for publication bias. Rev Bras Psiquitar. 2016 Jul-Sep;38(3):247–54. doi: 10.1590/1516-4446-2016-1915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Pereira DS, Mateo EC, de Queiroz BZ, et al. TNF-alpha, IL6, and IL10 polymorphisms and the effect of physical exercise on inflammatory parameters and physical performance in elderly women. Age (Dordrecht, Netherlands) 2013 Dec;35(6):2455–63. doi: 10.1007/s11357-013-9515-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Lucassen PJ, Meerlo P, Naylor AS, et al. Regulation of adult neurogenesis by stress, sleep disruption, exercise and inflammation: Implications for depression and antidepressant action. European neuropsychopharmacology. 2010 Jan;20(1):1–17. doi: 10.1016/j.euroneuro.2009.08.003. [DOI] [PubMed] [Google Scholar]
  • 132.Eyre H, Baune BT. Neuroimmunological effects of physical exercise in depression. Brain Behav Immun. 2012 Feb;26(2):251–66. doi: 10.1016/j.bbi.2011.09.015. [DOI] [PubMed] [Google Scholar]
  • 133.Gleeson M, Bishop NC, Stensel DJ, et al. The anti-inflammatory effects of exercise: mechanisms and implications for the prevention and treatment of disease. Nat Rev Immunol. 2011 Aug 05;11(9):607–15. doi: 10.1038/nri3041. [DOI] [PubMed] [Google Scholar]
  • 134.Martinsen EW, Hoffart A, Solberg O. Comparing aerobic with nonaerobic forms of exercise in the treatment of clinical depression: a randomized trial. Compr Psychiatry. 1989 Jul-Aug;30(4):324–31. doi: 10.1016/0010-440x(89)90057-6. [DOI] [PubMed] [Google Scholar]
  • 135.Dimeo F, Bauer M, Varahram I, et al. Benefits from aerobic exercise in patients with major depression: a pilot study. Br J Sports Med. 2001 Apr;35(2):114–7. doi: 10.1136/bjsm.35.2.114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Trivedi MH, Greer TL, Grannemann BD, et al. Exercise as an augmentation strategy for treatment of major depression. J Psychiatr Pract. 2006 Jul;12(4):205–13. doi: 10.1097/00131746-200607000-00002. [DOI] [PubMed] [Google Scholar]
  • 137.Trivedi MH, Greer TL, Church TS, et al. Exercise as an augmentation treatment for nonremitted major depressive disorder: a randomized, parallel dose comparison. J Clin Psychiatry. 2011 May;72(5):677–84. doi: 10.4088/JCP.10m06743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Schuch FB, Vancampfort D, Rosenbaum S, et al. Exercise improves physical and psychological quality of life in people with depression: A meta-analysis including the evaluation of control group response. Psychiatry Res. 2016 Jul 30;241:47–54. doi: 10.1016/j.psychres.2016.04.054. [DOI] [PubMed] [Google Scholar]
  • 139.Ng F, Dodd S, Berk M. The effects of physical activity in the acute treatment of bipolar disorder: a pilot study. J Affect Disord. 2007 Aug;101(1–3):259–62. doi: 10.1016/j.jad.2006.11.014. [DOI] [PubMed] [Google Scholar]
  • 140.Knubben K, Reischies FM, Adli M, et al. A randomised, controlled study on the effects of a short-term endurance training programme in patients with major depression. Br J Sports Med. 2007 Jan;41(1):29–33. doi: 10.1136/bjsm.2006.030130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Bercik P, Verdu EF, Foster JA, et al. Chronic gastrointestinal inflammation induces anxiety-like behavior and alters central nervous system biochemistry in mice. Gastroenterology. 2010 Dec;139(6):2102–2112.e1. doi: 10.1053/j.gastro.2010.06.063. [DOI] [PubMed] [Google Scholar]
  • 142.Bravo JA, Dinan TG, Cryan JF. Alterations in the central CRF system of two different rat models of comorbid depression and functional gastrointestinal disorders. Int J Neuropsychopharmacol. 2011 Jun;14(5):666–83. doi: 10.1017/s1461145710000994. [DOI] [PubMed] [Google Scholar]
  • 143.Bercik P, Collins SM, Verdu EF. Microbes and the gut-brain axis. Neurogastroenterol Motil. 2012 May;24(5):405–13. doi: 10.1111/j.1365-2982.2012.01906.x. [DOI] [PubMed] [Google Scholar]
  • 144.Wallace CJK, Milev R. Erratum to: The effects of probiotics on depressive symptoms in humans: a systematic review. Ann Gen Psychiatry. 2017;16:18. doi: 10.1186/s12991-017-0141-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Ait-Belgnaoui A, Durand H, Cartier C, et al. Prevention of gut leakiness by a probiotic treatment leads to attenuated HPA response to an acute psychological stress in rats. Psychoneuroendocrinology. 2012 Nov;37(11):1885–95. doi: 10.1016/j.psyneuen.2012.03.024. [DOI] [PubMed] [Google Scholar]
  • 146.Luo J, Wang T, Liang S, et al. Ingestion of Lactobacillus strain reduces anxiety and improves cognitive function in the hyperammonemia rat. Sci China Life Sci. 2014 Mar;57(3):327–35. doi: 10.1007/s11427-014-4615-4. [DOI] [PubMed] [Google Scholar]
  • 147.Ait-Belgnaoui A, Colom A, Braniste V, et al. Probiotic gut effect prevents the chronic psychological stress-induced brain activity abnormality in mice. Neurogastroenterol Motil. 2014 Apr;26(4):510–20. doi: 10.1111/nmo.12295. [DOI] [PubMed] [Google Scholar]
  • 148.Desbonnet L, Garrett L, Clarke G, et al. Effects of the probiotic Bifidobacterium infantis in the maternal separation model of depression. Neuroscience. 2010 Nov 10;170(4):1179–88. doi: 10.1016/j.neuroscience.2010.08.005. [DOI] [PubMed] [Google Scholar]
  • 149.Marin IA, Goertz JE, Ren T, et al. Microbiota alteration is associated with the development of stress-induced despair behavior. Scientific reports. 2017 Mar 07;7:43859. doi: 10.1038/srep43859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Messaoudi M, Violle N, Bisson JF, et al. Beneficial psychological effects of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in healthy human volunteers. Gut microbes. 2011 Jul-Aug;2(4):256–61. doi: 10.4161/gmic.2.4.16108. [DOI] [PubMed] [Google Scholar]
  • 151.Rao AV, Bested AC, Beaulne TM, et al. A randomized, double-blind, placebo-controlled pilot study of a probiotic in emotional symptoms of chronic fatigue syndrome. Gut pathogens. 2009 Mar 19;1(1):6. doi: 10.1186/1757-4749-1-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Huang R, Wang K, Hu J. Effect of Probiotics on Depression: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Nutrients. 2016 Aug 06;8(8) doi: 10.3390/nu8080483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Munoz-Elias G, Woodbury D, Black IB. Marrow stromal cells, mitosis, and neuronal differentiation: stem cell and precursor functions. Stem cells. 2003;21(4):437–48. doi: 10.1634/stemcells.21-4-437. [DOI] [PubMed] [Google Scholar]
  • 154.Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999 Apr 02;284(5411):143–7. doi: 10.1126/science.284.5411.143. [DOI] [PubMed] [Google Scholar]
  • 155.Salem H, Rocha NP, Colpo GD, et al. Moving from the Dish to the Clinical Practice: A Decade of Lessons and Perspectives from the Pre-Clinical and Clinical Stem Cell Studies for Alzheimer’s Disease. J Alzheimers Dis : JAD. 2016 Jul 2; doi: 10.3233/JAD-160250. [DOI] [PubMed] [Google Scholar]
  • 156.Colpo GD, Ascoli BM, Wollenhaupt-Aguiar B, et al. Mesenchymal stem cells for the treatment of neurodegenerative and psychiatric disorders. An Acad Bras Cienc. 2015 Aug;87(2 Suppl):1435–49. doi: 10.1590/0001-3765201520140619. [DOI] [PubMed] [Google Scholar]
  • 157*.Tfilin M, Sudai E, Merenlender A, et al. Mesenchymal stem cells increase hippocampal neurogenesis and counteract depressive-like behavior. Mol Psychiatry. 2010 Dec;15(12):1164–75. doi: 10.1038/mp.2009.110. Original paper showing the antidepressant effects of MSCs in an animal model of depression. [DOI] [PubMed] [Google Scholar]
  • 158.Coquery N, Blesch A, Stroh A, et al. Intrahippocampal transplantation of mesenchymal stromal cells promotes neuroplasticity. Cytotherapy. 2012 Oct;14(9):1041–53. doi: 10.3109/14653249.2012.694418. [DOI] [PubMed] [Google Scholar]
  • 159.Wiedlocha M, Marcinowicz P, Krupa R, et al. Effect of antidepressant treatment on peripheral inflammation markers - A meta-analysis. Prog Neuropsychopharmacol Biol Psychiatry. 2017 Apr 23; doi: 10.1016/j.pnpbp.2017.04.026. [DOI] [PubMed] [Google Scholar]
  • 160.Rapaport MH, Manji HK. The effects of lithium on ex vivo cytokine production. Biol Psychiatry. 2001 Aug 01;50(3):217–24. doi: 10.1016/s0006-3223(01)01144-1. [DOI] [PubMed] [Google Scholar]
  • 161.Bosetti F, Rintala J, Seemann R, et al. Chronic lithium downregulates cyclooxygenase-2 activity and prostaglandin E(2) concentration in rat brain. Mol Psychiatry. 2002;7(8):845–50. doi: 10.1038/sj.mp.4001111. [DOI] [PubMed] [Google Scholar]
  • 162.Himmerich H, Schonherr J, Fulda S, et al. Impact of antipsychotics on cytokine production in-vitro. J Psychiatr Res. 2011 Oct;45(10):1358–65. doi: 10.1016/j.jpsychires.2011.04.009. [DOI] [PubMed] [Google Scholar]
  • 163.Aan Het Rot M, Zarate CA, Jr, Charney DS, et al. Ketamine for depression: where do we go from here? Biol Psychiatry. 2012 Oct 01;72(7):537–47. doi: 10.1016/j.biopsych.2012.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Yang C, Hong T, Shen J, et al. Ketamine exerts antidepressant effects and reduces IL-1beta and IL-6 levels in rat prefrontal cortex and hippocampus. Exp Ther Med. 2013 Apr;5(4):1093–1096. doi: 10.3892/etm.2013.930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Dale O, Somogyi AA, Li Y, et al. Does intraoperative ketamine attenuate inflammatory reactivity following surgery? A systematic review and meta-analysis. Anesth Analg. 2012 Oct;115(4):934–43. doi: 10.1213/ANE.0b013e3182662e30. [DOI] [PubMed] [Google Scholar]
  • 166.Wang N, Yu HY, Shen XF, et al. The rapid antidepressant effect of ketamine in rats is associated with down-regulation of pro-inflammatory cytokines in the hippocampus. Ups J Med Sci. 2015;120(4):241–8. doi: 10.3109/03009734.2015.1060281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Zunszain PA, Horowitz MA, Cattaneo A, et al. Ketamine: synaptogenesis, immunomodulation and glycogen synthase kinase-3 as underlying mechanisms of its antidepressant properties. Mol Psychiatry. 2013 Dec;18(12):1236–41. doi: 10.1038/mp.2013.87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Walker AK, Budac DP, Bisulco S, et al. NMDA receptor blockade by ketamine abrogates lipopolysaccharide-induced depressive-like behavior in C57BL/6J mice. Neuropsychopharmacology. 2013 Aug;38(9):1609–16. doi: 10.1038/npp.2013.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Kronfol Z, Hamdan-Allen G, Goel K, et al. Effects of single and repeated electroconvulsive therapy sessions on plasma ACTH, prolactin, growth hormone and cortisol concentrations. Psychoneuroendocrinology. 1991;16(4):345–52. doi: 10.1016/0306-4530(91)90020-t. [DOI] [PubMed] [Google Scholar]
  • 170.Fluitman SB, Heijnen CJ, Denys DA, et al. Electroconvulsive therapy has acute immunological and neuroendocrine effects in patients with major depressive disorder. J Affect Disord. 2011 Jun;131(1–3):388–92. doi: 10.1016/j.jad.2010.11.035. [DOI] [PubMed] [Google Scholar]
  • 171.Lehtimaki K, Keranen T, Huuhka M, et al. Increase in plasma proinflammatory cytokines after electroconvulsive therapy in patients with depressive disorder. J ECT. 2008 Mar;24(1):88–91. doi: 10.1097/YCT.0b013e3181571abb. [DOI] [PubMed] [Google Scholar]
  • 172.Hestad KA, Tonseth S, Stoen CD, et al. Raised plasma levels of tumor necrosis factor alpha in patients with depression: normalization during electroconvulsive therapy. J ECT. 2003 Dec;19(4):183–8. doi: 10.1097/00124509-200312000-00002. [DOI] [PubMed] [Google Scholar]
  • 173.Freire TFV, Rocha NSD, Fleck MPA. The association of electroconvulsive therapy to pharmacological treatment and its influence on cytokines. J Psychiatr Res. 2017 Sep;92:205–211. doi: 10.1016/j.jpsychires.2017.05.004. [DOI] [PubMed] [Google Scholar]
  • 174.Brunoni AR, Ferrucci R, Fregni F, et al. Transcranial direct current stimulation for the treatment of major depressive disorder: a summary of preclinical, clinical and translational findings. Prog Neuropsychopharmacol Biol Psychiatry. 2012 Oct 01;39(1):9–16. doi: 10.1016/j.pnpbp.2012.05.016. [DOI] [PubMed] [Google Scholar]
  • 175.Nitsche MA, Cohen LG, Wassermann EM, et al. Transcranial direct current stimulation: State of the art 2008. Brain Stimul. 2008 Jul;1(3):206–23. doi: 10.1016/j.brs.2008.06.004. [DOI] [PubMed] [Google Scholar]
  • 176.Spezia Adachi LN, Caumo W, Laste G, et al. Reversal of chronic stress-induced pain by transcranial direct current stimulation (tDCS) in an animal model. Brain Res. 2012 Dec 13;1489:17–26. doi: 10.1016/j.brainres.2012.10.009. [DOI] [PubMed] [Google Scholar]
  • 177.Brunoni AR, Machado-Vieira R, Zarate CA, et al. Cytokines plasma levels during antidepressant treatment with sertraline and transcranial direct current stimulation (tDCS): results from a factorial, randomized, controlled trial. Psychopharmacology. 2014 Apr;231(7):1315–23. doi: 10.1007/s00213-013-3322-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Benabid AL, Benazzous A, Pollak P. Mechanisms of deep brain stimulation. Mov Disord. 2002;17(Suppl 3):S73–4. doi: 10.1002/mds.10145. [DOI] [PubMed] [Google Scholar]
  • 179.Lozano AM, Giacobbe P, Hamani C, et al. A multicenter pilot study of subcallosal cingulate area deep brain stimulation for treatment-resistant depression. J Neurosurg. 2012 Feb;116(2):315–22. doi: 10.3171/2011.10.jns102122. [DOI] [PubMed] [Google Scholar]
  • 180.Calleja-Castillo JM, De La Cruz-Aguilera DL, Manjarrez J, et al. Chronic deep brain stimulation of the hypothalamic nucleus in wistar rats alters circulatory levels of corticosterone and proinflammatory cytokines. Clin Dev Immunol. 2013;2013:698634. doi: 10.1155/2013/698634. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES