Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2021

Open Access 01.12.2021 | Review

On inflammatory hypothesis of depression: what is the role of IL-6 in the middle of the chaos?

verfasst von: Elnaz Roohi, Nematollah Jaafari, Farshad Hashemian

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2021

Abstract

Many patients with major depressive disorder (MDD) are reported to have higher levels of multiple inflammatory cytokines including interleukin 6 (IL-6). Recent studies both pre-clinical and clinical have advocated for the functional role of IL-6 in development of MDD and suggested a great potential for targeting this cytokine to open new avenues in pharmacotherapy of depression. The purpose of the present narrative review was to provide an integrated account of how IL-6 may contribute to development of depression. All peer-reviewed journal articles published before July 2020 for each area discussed were searched by WOS, PubMed, MEDLINE, Scopus, Google Scholar, for original research, review articles, and book chapters. Publications between 1980 and July 2020 were included. Alterations in IL-6 levels, both within the periphery and the brain, most probably contribute to depression symptomatology in numerous ways. As IL-6 acts on multiple differing target tissues throughout the body, dysregulation of this particular cytokine can precipitate a multitude of events relevant to depression and blocking its effects can prevent further escalation of inflammatory responses, and potentially pave the way for opening new avenues in diagnosis, treatment, and prevention of this debilitating disorder.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ACC
Anterior cingulate cortex
ARDS
Acute respiratory distress syndrome
BBB
Blood-brain barrier
CNS
Central nervous system
COVID-19
Coronavirus disease 2019
CRS
Cytokine release syndrome
CTRA
Conserved transcriptional response to adversity
DMARDs
Disease-modifying anti-rheumatic drugs
DSM-5
Diagnostic and Statistical Manual of Mental Disorder, 5th Edition
ECT
Electroconvulsive therapy
Gp130
Glycoprotein 130
GPCRs
G-protein-coupled receptors
HPA
Hypothalamic-pituitary-adrenal
IDO
Indoleamine 2,3 dioxygenase
IFN
Interferon
IL
Interleukin
IL-6R
IL-6 receptor
JAK/STAT
Janus kinase/signal transducer and activator of transcription
KYN
Kynurenine
MDD
Major depressive disorder
MAPK
Mitogen-activated protein kinase
mRNA
Messenger RNA
NF-κB
Nuclear factor kappa-light-chain-enhancer of activated B cells
PFC
Prefrontal cortex
RA
Rheumatoid arthritis
SARS-CoV-2
severe acute respiratory syndrome coronavirus 2
SERT
Serotonin transporter
STAT3
Signal transducer and activator of transcription 3
sIL-6R
Soluble IL-6R
TGF
Transforming growth factor
TH2
T helper type 2
TH1
T helper type 1
Th
T helper cell
TNF
Tumor necrosis factor
TSPO
Translocator protein
Treg
Regulatory T cells

Background

Major depressive disorder (MDD) is a leading cause of disability throughout the world with a global prevalence of 2.6–5.9% [1]. The total estimated number of people living with depression worldwide increased by 49.86% from 1990 to 2017 [2]. According to worldwide projections, MDD will be the single major cause of burden of all health conditions by 2030 [3]. MDD is characterized by periods of low mood, altered cognition, considerable functional burden including impaired occupational functioning and psychosocial disability [4]. Despite available pharmacotherapeutic options, 30–60% of patients with MDD are not responsive to available treatments [5] and the rate of remission of the disease is often < 50% [6], while recurrence rates are more than 85% within 10 years of a depressive episode, and average about ≥ 50% within 6 months of assumed clinical remission [4]. Indeed, there exists no compelling evidence that current treatments are capable of disease modification in MDD patients. Thus, therapeutic deficiency in treatment outcomes reflects the demand for revitalizing psychiatric therapeutics with novel pharmacotherapeutic options that engage non-monoaminergic molecular targets.
A large body of evidence suggests that inflammation has central role in pathogenesis of MDD [713]. However, the exact mechanisms underlying inflammation-induced depression are not completely elucidated [3]. Historically, the “monoamine-depletion hypothesis” has been the main proposed pathophysiology [14]; nevertheless, this hypothesis alone cannot fully account for pathogenesis of MDD [15, 16]. In recent years, “inflammatory hypothesis” has been proposed [17]. However, it is noteworthy that it was probably in the early 1990s that for the first time, possible relationships between the peripheral immune system and major depression was studied [18]. Maes et al. (1992) established immune cell profile of patients with depression and advocated for the existence of a systemic immune activation during major depressive disorder [19]. Moreover, correlations between IL-6 activity, acute phase proteins, and hyperactivity of the hypothalamic-pituitary-adrenal (HPA) axis were suggested in severe depression [20].
Most proximally, inflammation is regulated by expression of immune response genes including interleukin (IL)-1B, tumor necrosis factor (TNF), and IL-6 which promote secretion of pro-inflammatory cytokines leading to systemic inflammation. Distally, inflammation is regulated in the brain where socio-environmental cues including possible threat are detected. This neuro-inflammatory link can activate the conserved transcriptional response to adversity (CTRA) before happening of a possible threat or bacterial infection. However, the negative aspect of central regulation of systemic inflammation is that it can give social and foreseen dangers (including those that have not yet occurred or may never actually happen) the ability to activate the CTRA in the absence of actual physical danger. Under normal conditions, CTRA-related inflammatory activity is downregulated by the HPA axis via the production of cortisol. Nevertheless, when prolonged actual or perceived social threat or physical danger is present, glucocorticoid resistance can develop which leads to excessive inflammation that heightens a person’s risk for development of several disorders including MDD, especially if activation of these pathways is prolonged [21]. As mentioned above, the current understanding of MDD encloses not only alterations in neurotransmitters, but also changes in immune and endocrine functioning as well as neural circuits [22]. This broadened framework has just started to inform a wide array of novel, personalized therapeutics that are showcasing great promise in a new holistic approach to MDD [23].
Cytokines are implicated in pathogenesis of MDD [2430]. Risk factors of developing MDD include familial, developmental, psychological, and medical risk factors as well as molecular factors associated with genetics, epigenetics, gene expression, and also those related to the endocrine and the immune system [31, 32]. All these risk factors have been shown to be related with changes in cytokine production or signaling. In other words, cytokines are involved in almost every predisposing or precipitating risk factor associated with MDD [24]. Indeed, there is accumulating evidence in favor of involvement of pro-inflammatory cytokines in pathophysiology of depression [24, 29, 3336]. Various studies reported higher levels of multiple inflammatory markers including IL-6 in patients with MDD [3741]. Of all pro-inflammatory cytokines, changes in IL-6 serum levels have been reported as one of the most reproducible abnormalities in MDD [38].
The aim of the present narrative review is to elucidate the fundamentals, implications, challenges of cytokine research specifically IL-6 in major depressive disorder. This comprises of the following:
-) A Brief overview of cytokines
-) Cytokine categories according to immunological function.
-) IL-6 as a pleiotropic cytokine.
-) Brief overview of chemokines and their role in Depression.
-) Challenges of cytokine research in psychiatry.
-) IL-6 alterations in depression.
-) Effects of IL-6 on neurotransmitters’ synthesis, signaling, metabolism, and function.
-) Effects of IL-6 levels on brain morphology in depression.
-) Blockade of IL-6 and its receptor in the periphery as a potential therapeutic option in MDD.
-) Possible role of IL-6 together with gut microbiota in pathogenesis of depression.
-) Elevated levels of IL-6 in patients with COVID-19 infection.

Methods

The present article is a narrative review. All peer-reviewed journal articles published before July 2020 for each area discussed were searched by WOS, PubMed, MEDLINE, Scopus, Google Scholar, for original research, review articles, and book chapters. We selected articles on the basis of being comprehensive, innovative, and informative for an in-depth understanding and a critical debate on the topic. Publications between 1973 and 2020 were included.

A brief overview of cytokines

Cytokines are a broad category of released proteins that act as signaling molecules to regulate inflammation and cellular activities [24, 42]. They are produced by different immune cells (e.g., macrophages, lymphocytes, mast cells), parenchymal cells, endothelial and epithelial cells, fibroblasts, adipocytes, and stromal cells within the periphery [24, 43]. Additionally, they are produced by microglia, astrocytes, and neurons in the brain [44]. Cytokines from the periphery (peripherally produced cytokines) can exert influences on inflammatory processes in the brain [45, 46]. Indeed, they can enter blood-brain barrier (BBB) and affect the brain via humoral (accessing the brain through leaky secretions of the BBB such as choroid plexus), neural (through stimulation of primary afferent nerve fibers in the vagus nerve), and cellular (through stimulation of microglia by pre-inflammatory cytokines to produce monocyte chemottractant protein-1 and recruit monocytes to the brain) pathways [47]. Most cytokines function in their immediate microenvironment. Few of them are involved in paracrine signaling which indeed is fundamental to the control of an inflammatory response within a given tissue or organ and the activation of a coordinated immune response that involves multiple cell types [48]. Apart from navigating the immune system to defend the body from pathogens, cytokines have a modifying effect on neurotransmission [49].
It’s also noteworthy that the same cytokines can be produced by multiple cell types. For example, white blood cells, endothelium, fat cells, and other cells can produce TNF-α [50]. Additionally, one single cell can release different cytokines. For instance, T Helper type 2 (TH2) cells can produce IL-3, IL-4, IL-5, IL-6, and IL-13 [24]. Cytokines can have pleiotropic, redundant, synergistic, and antagonistic effects [51]. The phenomenon that a single cytokine can act on several different cell types is called pleiotropy [51]. For instance, IL-10 can activate TH2 cells and B cells, yet inhibit macrophages and T helper type 1 (TH1) cells. Thus, being immunostimulatory as well as being immunosuppressive [52]. Cytokines are redundant in their activity, i.e., similar functions can be exerted by different cytokines. For instance, interferon (IFN)-γ, IL-2, and TNF-α enhance cellular immunity and production of cytotoxic cell contacts [53]. Cytokines can also act synergistically, i.e., they can have combined effects when acting together. For instance, IL-3 and IL-4 amplify each other’s effects to induce growth, differentiation, and activation of mast cells in a synergistic manner [24]. Another phenomenon in cytokines signaling is antagonism. An example of cytokine antagonism is that cytokines of the IL-1 superfamily can antagonize IL-18 effects [54].

Cytokine categories according to immunological function

Four categories of cytokines are usually referred to in psychoimmunological literature. (1) TH1 cytokines (IL-2, IL-12, IFN-γ) which induce cytotoxic cell contacts. (2) TH2 cytokines (IL-4, (IL-5, IL-13) which lead to production of antibodies. (3) Pro-inflammatory cytokines (IL-1, IL-6, IL-8, IL-17, IL-21, IL-22, IFN-α, TNF-α) which further the progress of inflammation. (4) Anti-inflammatory cytokines (IL-10, transforming growth factor-beta (TGF)-β which are influenced by regulatory T cells and impede inflammatory process from escalating [24]. However, these categories are not distinct and it must be considered that cytokines can exert various effects on different cells and therefore, they may have pro- and also anti-inflammatory properties. For instance, IFN-α which has been listed as a pro-inflammatory cytokine can also have anti-inflammatory properties [55].

IL-6 as a pleiotropic cytokine

IL-6 was first identified as a differentiation factor for B cells which stimulates production of antibodies by activated B cells. Apart from regulation of acute inflammation, IL-6 is known to induce differentiation of B cells, and activation and population expansion of T cells [56]. Within the peripheral and central nervous system (CNS), IL-6 can act as a neuronal growth factor inducing neurite development and nerve regeneration [57]. IL-6 receptor (IL-6R) consists of the IL-6-binding chain which has two forms of transmembrane IL-6R and soluble IL-6R (sIL-6R) [58] and a gp130 signal-transducing chain [59]. Following binding to its receptor (IL-6R), IL-6 initiates to exert its multiple functions.
It is quite interesting that IL-6 exerts both pro- and anti-inflammatory properties [60, 61]. Indeed, its signaling is complex and can lead to both inflammatory and anti-inflammatory cascades depending upon the presence of either IL-6 receptor (IL-6R) or the membrane bound gp130 signal transducer and these are expressed at very different frequencies within specific cell type in the body [5]. Trans-signaling of IL-6, in which the soluble form of the IL-6 receptor (sIL-6R) is shed from the membrane bound receptors, is known to be pro-inflammatory [62]. The sIL-6R binds to IL-6 and is transported to any cell type on which gp130 is expressed [63]. While most soluble receptors (e.g., soluble receptor for TNFα) result in antagonistic action by competing for the ligand, the sIL-6R is agonistic and increases the types of cells through which IL-6 can signal. Additionally, IL-6 engages in classical signaling which is anti-inflammatory [63] and occurs through binding of IL-6 to the membrane bound cell surface receptor. Classical signaling of IL-6 solely occurs on some subsets of T cells, neutrophils and monocytes megakaryocytes, and hepatocytes [64]. In both classical and trans-signaling, the IL-6/IL-6R/gp130 complex uses two pathways to activate intracellular signaling namely the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway and the mitogen-activated protein kinase (MAPK) pathway [5].
Indeed, IL-6 has been mostly regarded as having pro-inflammatory properties; however, it has many anti-inflammatory functions which are necessary for resolution of inflammation [65]. For instance, IL-6 inhibits activity of the transcription factor named nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and expression of the chemokine receptor on dendritic cells which is needed for recruiting these cells to lymphoid tissues; thus, involving in resolution of inflammation [66]. Research findings showed that IL-6 has a crucial role in regulation of T helper17 (Th17)/regulatory T (Treg) cells [67]. In the presence of TGF-β, IL-6 is a vital signal for differentiation of naive T cells into Th17 cells which in turn are implicated in induction of autoimmune diseases [68, 69], and result in local tissue injury in chronic inflammatory disorders [70]. On the contrary, IL-6 can strongly inhibit the TGF-β-induced differentiation of Treg cells which in turn results in inhibition of autoimmunity and protects against tissue damage [71]. Functional dichotomy of IL-6 indicates that it may be responsible for maintaining the balance between pro- and anti-inflammatory responses, while having tissue-specific properties at the periphery and in the CNS [72].

Brief overview of chemokines and their role in depression

Chemokines are small chemotactic cytokines that are identified to have significant roles in migration of immune cells, induction of direct chemotaxis, and propagation of inflammatory responses [73]. They are classified into four sub-families based on their structural criteria (i.e., the number and spacing of their two N-terminals, disulfide bonding participating cysteine residues). These four subfamilies include CXC, CC, C, and CX3C [74]. Furthermore, they can be categorized according to their biological activity, namely, the maintenance of homeostasis and the induction of inflammation. There are also chemokines which have dual functionality [75].
These small chemotactic cytokines are known to be secreted in response to inflammatory cytokines; thereafter, selectively recruiting lymphocytes, monocytes, and neutrophil-inducing chemotaxis by activating G-protein-coupled receptors (GPCRs) [76]. A growing body of evidence suggests that chemotactic cytokines are implicated in neurobiological processes relevant to psychiatric disorders such as synaptic transmission and plasticity, neuroglia communication, and neurogenesis [77]. Indeed, disruption of any of the mentioned functions which may take place by activation of the inflammatory response system has consistently been found to be relevant in pathogenesis of depression [73].
There are indeed both pre-clinical and clinical evidence in support of linking alterations in the chemokine network to depressive behavior [73]. In an animal model of depression, namely prenatal stressed rats, levels of CCL2, and CXCL12 chemokines were found to be upregulated in the hippocampus and prefrontal cortex, which was indeed suggestive of excessive microglial activation [78]. Additionally, Trojan et al. (2017) investigated the modulatory properties of chronic administration of anti-depressants on the chemokines. According to their results, chronic administration of anti-depressants has been shown to normalize the prenatal stress-induced behavioral disturbances together with the observed alterations in CXCL12 and their receptor. Indeed, they concluded that alterations of CXCL12 and their receptor and at less extend changes in CX3CL1–CX3CR1 expression will probably be normalized following chronic treatment with anti-depressants [79].
Moreover, several clinical studies found correlations between elevated levels of circulating inflammatory chemokines and depressive symptoms in patients with major depressive disorder [73, 8083]. According to the results of a comprehensive meta-analysis, peripheral concentrations of a number of chemokines including CCL2, CCL3, CCL4, CCL11, CXCL4, CXCL7, and CXCL8 can potentially discriminate between individuals with depression and those without [84]. Additionally, Ślusarczyk et al. (2016) provided a comprehensive account of the role of chemokines in processes underlying depressive disorder [85].

Challenges of cytokine research in psychiatry

There are some difficulties faced by researchers in conducting cytokine research in psychiatry. The major problem seems to be heterogeneity of the obtained results. In other words, research outcomes are conflicting and challenging to interpret [24]. Moreover, research in this area is largely based on measurement of cytokine levels in the periphery and it is not completely clear how serum or plasma levels of cytokines reflect the situation in the brain [47]. Compellingly, results of studies that examined both peripheral and CFS levels of IL-6 found no correlations between the mentioned measures; thus, suggesting that peripheral levels of IL-6 may not directly reflect central IL-6 levels [86, 87].
It is also noteworthy that some environmental, social, biological, and medical factors may influence peripheral cytokine changes. For instance, one of the characteristics of obesity is chronic inflammation with the increased circulating levels of cytokines [88, 89]. Indeed, adipose tissue is reported to build up and activate lymphocytes and macrophages that secrete inflammatory factors [89, 90]. Interestingly, obese people show behavioral symptoms such as MDD and cognitive dysfunction at an increased rate in comparison with the general population [91, 92]. Therefore, one may argue that alterations in cytokine levels are somehow unspecific [24]. Additionally, there is a considerable overlap in cytokine values between patients in the acute phase of depression, patients in remission, and patients who are recovered [24]. Although the use of cytokines as potential biomarkers of depression has been discussed frequently in various studies, cytokine changes have been reported in other psychiatric disorders as well. For instance, increased levels of pro-inflammatory cytokines have been reported in generalized anxiety disorder [9395], obsessive-compulsive disorder [96], posttraumatic stress disorder [97, 98], and sleep disorder [99].
Moreover, cytokine levels change during pharmacotherapy of depression. Indeed, it has been suggested that treatment with anti-depressants can potentially lead to alteration in peripheral levels of cytokines. According to the results of a meta-analysis, anti-depressants, overall, cause decrease in peripheral levels of IL-6, IL-10, and TNF-α [100]. However, anti-psychotics which are used in psychotic depression, especially those with the highest risk of weight gain (e.g., clozapine and olanzapine), cause significant increase in the blood levels of pro-inflammatory cytokine [101]. Additionally, mood stabilizers such as lithium and carbamazepine have also been linked with an increase in the peripheral levels of cytokines [102]. In sum, as cytokine signaling often exhibits pleiotropic, redundant, synergistic, and antagonistic effects, it seems to be advisable to consider all cytokines that work together or against each other and therefore, take into account the whole range of cytokines instead of a single one.

On the role of interleukin-6 in depression

IL-6 alterations in depression

A growing body of evidence suggests that IL-6 has a crucial role in pathogenesis of depression [3] and is the most consistently increased cytokine in blood samples of MDD patients [38, 103]. The first promising evidence for the role of IL-6 in occurrence of depression is most probably provided by a longitudinal study in which children with higher circulating levels of IL-6 at age 9 were found to be at a 10% greater risk of developing MDD by age 18, compared to the general population or children with low IL-6 levels. Indeed, the researchers concluded that inflammation and high IL-6 levels possibly predate the occurrence of depression [104]. Another evidence for potential role of IL-6 in depression is that peripheral levels of IL-6 were found to be positively correlated with symptom severity in anti-depressant non-responders [105].
Stress-based preclinical models of depression showed that IL-6 levels are increased following the onset of depression-associated behaviors. Rodents who were exposed to chronic mild stress exhibited anhedonia and elevated circulating levels of pro-inflammatory cytokines including IL-6 [106, 107]. Moreover, in another study on male Wistar rats, serum levels of pro-inflammatory cytokines including IL-6 were reported to be higher in acute and restraint stress compared to non-stressed rats [108]. It is also noteworthy that some studies reported no significant alteration in peripheral levels of IL-6 in chronic mild stress models of depression [109]. Nevertheless, they reported elevated CNS levels of other inflammatory markers which probably reflects a time-dependent shift from peripheral to central cytokine activation or potential transport of the peripheral cytokines into CNS [109]. Another promising evidence was provided by studies on IL-6 knockout mice. Indeed, they were reported to be resistant to the development of depression-like phenotype following long-term light deprivation in the constant darkness, proposing a functional role for IL-6 in stress susceptibility [110]. Moreover, Ślusarczyk et al. (2015) found evidence for the role of prenatal stress as a priming factor that could exhibit effects on microglial cells and consequently lead to depressive-like disturbances in adult rat offspring. According to their results, the release of pro-inflammatory cytokines including IL-6 is enhanced in microglia obtained from prenatally stressed animals compared to control animals [78].
In fact, not every individual exposed to prolonged or acute stress develops a psychiatric disorder [111]. According to previous research, vulnerability to repeated social defeat stress is predicted by differences in IL-6 levels in the innate peripheral immune system [112]. Following induction of social defeat stress, two thirds of mice were reported to show depression-like behavior measured by social avoidance, anhedonia, circadian system disruptions, and metabolic changes [113] together with elevated activation of pro-inflammatory cytokines such as IL-6 [112]. Indeed, higher degrees of elevation in peripheral IL-6 levels of susceptible mice were reported in comparison with resilient mice. Moreover, it was found that this increase occurs within 20 min of the first social defeat. Interestingly, mice that later became susceptible had higher number of leukocytes and those leukocytes produced more levels of IL-6 following stimulation via LPS ex vivo [112]. Additionally, studies with non-social stress-based models found evidence for the functional role of IL-6 in the development of stress susceptibility. In these models, animals were exposed to a controllable or uncontrollable stress (e.g., shock), and their ability to actively escape a subsequent stressor was measured. According to the results, 20% of animals who were exposed to uncontrollable stress were found susceptible and developed learned helplessness and the rest were found to be resilient. Interestingly, susceptible animals showed elevated levels of peripheral IL-6 together with anhedonia in contrast to resilient animals [114].
Clinical studies have also revealed that patients with MDD have increased levels of plasma and serum concentrations of pro-inflammatory cytokines including IL-6 in comparison with healthy controls [24, 100, 115, 116]. It should be noted that three meta-analyses verified increased peripheral IL-6 levels in MDD patients compared to healthy volunteers [38, 116, 117]. Nevertheless, there are also studies reporting no significant differences in IL-6 levels in MDD patients compared to healthy volunteers [118]. However, one may argue that different subtypes of depression and certain depressive symptoms should be taken into account. For instance, Rudolf et al. (2014) compared IL-6 levels among patients with atypical and typical depression and healthy controls. According to their results, IL-6 levels were significantly increased in patients with atypical depression and not in typical MDD patients compared to healthy controls [119]. Additionally, Rush et al. (2016) studied peripheral levels of IL-6 and TGF-β in 55 melancholic depressed patients. They were found to have significantly higher baseline IL-6 levels compared to healthy controls. Moreover, these elevated levels of IL-6 did not normalize following electroconvulsive therapy (ECT) [120]. A recent systematic review conformed Rush et al.’s results. In the mentioned review, authors found that peripheral IL-6 levels are increased in patients with melancholic depression in comparison with controls [121]. Moreover, Maes et al. (1997) examined serum levels of IL-6 and IL-1 receptor antagonist in patients with chronic, treatment resistant depression both before and after subchronic treatment with anti-depressants. According to their results, subchronic treatment with anti-depressants had no significant impact on serum levels of IL-6; nevertheless, it decreased serum soluble IL-6R levels significantly [122].

Effects of IL-6 on neurotransmitters’ synthesis, signaling, metabolism, and function

The effects of cytokines on neurotransmitters have been studied extensively [49, 123]. Cytokines and their signaling pathways (e.g., p38 mitogen activated protein kinase) are reported to exhibit significant impacts on metabolism of multiple neurotransmitters such as serotonin, dopamine, and glutamate; thus, influencing their synthesis, release, and reuptake [49]. Indeed, cytokines can decrease synthesis of serotonin via activating the enzyme indoleamine 2,3 dioxygenase (IDO) which breaks the precursor of serotonin (i.e., tryptophan) to kynurenine (KYN) instead of metabolizing tryptophan to serotonin; thus, leading to serotonin depletion [3]. The process of serotonin depletion has been long associated with major depression [124, 125]. Moreover, cytokines can modulate serotonin signaling via elevating the expression and function of monoamine transporters. These transporters are known to re-uptake serotonin [126, 127].
IL-6 is known to influence neurotransmission by modulating the behavioral output of the brain; however, the exact mechanism is unknown. A previous study showed that IL-6 directly controls the levels of serotonin transporter (SERT) and therefore influences serotonin reuptake. Indeed, the researchers concluded that IL6-induced modulation of serotonergic neurotransmission through the signal transducer and activator of transcription 3 (STAT3) signaling pathway contributes to the role of IL6 in depression [128]. The activity of SERT forms serotonergic transmission which is implicated in depressive behavioral changes and pathophysiology of the disease [129]. By intensifying dopaminergic and serotonergic turnover in hippocampus and frontal cortex, IL-6 influences neurotransmission of catecholamines [130]. It seems that noradrenaline is not affected by IL-6; however, noradrenaline itself can induce expression of IL-6 in glial cells [131]. IL-6 together with other pro-inflammatory cytokines can activate kinurenine pathway which is involved in glutamatergic neurotransmission [132].

Effects of IL-6 levels on brain morphology in depression

Previous studies showed that elevated levels of pro-inflammatory cytokines such as IL-6 may affect neurogenesis [133] and neural plasticity [134]. Imaging studies have shown that specific brain regions such as basal ganglia (which is involved in motor activity and motivation), the dorsal anterior cingulate cortex (ACC) (which has a central role in generation of anxiety), and the subgenual ACC (which is known to be involved in the development of depression) are influenced by cytokines [135, 136]. Additionally, high IL-6 expression levels demonstrated neuropathologic manifestations including neurodegeneration [137, 138].
There are many studies in which positron emission tomography (PET) has been applied to test translocator protein (TSPO) binding, a marker of neuroinflammation, in order to study neuroinflammatory hypothesis of depression [139143]. According to their results, neuroinflammation was present in various regions of the brain (e.g., neocortical grey matter, frontal cortex, prefrontal cortex, anterior cingulate cortex, insula, temporal cortex) as well as the hippocampus [139143].
In a recent study, Kakeda et al. (2018) evaluated possible relationship between serum levels of IL-1β, IL-6, IFN-γ, and TNFα and brain morphology in terms of brain cortical thinning and hippocampal subfield volumes during the first depressive episode in drug-naïve patients with MDD using a whole-brain SBM analysis. They found a significant inverse correlation between prefrontal cortex (PFC) thickness and serum IL-6 level in MDD patients. Indeed, high serum levels of IL-6 were correlated with reduced left subiculum and right CA1, CA3, CA4, GC-DG, subiculum, and whole hippocampus volumes in MDD patients. Additionally, thickness of the superior frontal and medial orbitofrontal cortices in patients with depression was significantly decreased compared to healthy controls. Since PFC contains high concentrations of IL-6 receptors, IL-6 mediated neurotoxicity might happen under conditions in which high serum IL-6 levels are present (i.e., early stages of MDD). Consequently, the authors advocated that the neuroinflammatory status in the early stage of MDD is associated with changes in the brain gray matter and IL-6 probably plays a key role in the morphological changes observed in the PFC during early stages of the disease. It is also noteworthy that serum IL-6 was the only cytokine among the tested cytokines that showed significant differences between the patients and controls in their study. Indeed, serum IL-6 levels were found to be significantly higher in MDD patients than in healthy controls [144]. In another study, Frodl et al. (2012) investigated possible effects of changes in the glucocorticoid and inflammatory systems on hippocampal volumes in patients with MDD. According to their results, MDD patients showed increased IL-6 levels and smaller hippocampal volumes compared to healthy controls. Positive effects of messenger RNA (mRNA) expression of glucocorticoid-inducible genes and further inverse effects of IL-6 concentration, on hippocampal volumes were also reported. Thus, they concluded that increased expression of IL-6 can probably predict decreased hippocampal volume [145].
As already mentioned, there is considerable amount of evidence regarding the central role of the highly plastic, stress-sensitive hippocampal region in pathogenesis of depression [146]. Indeed, grey-matter structures, including the hippocampus are vulnerable to atrophy in depression [147, 148]. Hippocampal volume reductions are most probably the result of remodeling of key cellular elements, involving retraction of dendrites, loss of glial cells, and decreased neurogenesis in the dentate gyros [149]. Factors underlying this cellular remodeling are known to be stress-induced increased levels of glucocorticoids, which are implicated in decreased neurogenesis [150]. Moreover, increased activity of the HPA axis resulting in decreased levels of glucocorticoids combined with resistance to glucocorticoid-induced negative feedback control is commonly observed in depression [151]. This dysregulation of glucocorticoid secretion along with the increased activity of excitatory neurotransmitters can potentially lead to cellular remodeling (which can be reversible) and hippocampal neurons cell death in patients with depression [152]. Since hippocampus has been identified to have a role in negative feedback inhibition of glucocorticoids, remodeling or neuronal damage may lead to less efficient inhibitory control of the corticotrophin-releasing hormone, resulting in elevated amounts of circulating glucocorticoids and further damage of the hippocampal neurons [153]. Taken together, it seems that further studies are required to elucidate the physiological mechanisms in which IL-6 might exert changes in the brain grey matter. A brief overview of the effects of cytokines including IL-6 on brain morphology is shown in Fig. 1.

Blockade of IL-6 and its receptor in the periphery as a potential therapeutic option in MDD

Growing body of evidence suggests that abnormalities in the immune system are most probably relevant to pathogenesis and potential novel treatment of psychiatric disorders. Previous studies showed that alterations in the peripheral IL-6 levels might contribute to depressive-like behavior in animal studies [3, 112, 114, 154]. Moreover, IL-6 knockout mice showed resistance to development of depressive-like behavior [155] which gives further evidence for possible role of IL-6 pathogenesis of depression. High peripheral levels of IL-6 are even more apparent in patients with treatment-resistant depression. Additionally, correlations have been found between decrease in IL-6 levels and alleviation of depressive symptoms in patients who were responsive to the pharmacotherapy [156]. Moreover, results of a study on 222 stroke patients indicated significant associations between IL-6 periphery levels and development of MDD within 2 weeks and at 1 year following stroke. Furthermore, significant correlations were found between statin use and IL-6 on the presence of a depressive disorder at the 1st year. Indeed, preventive effects of treatment with statins (which are known to possess anti-inflammatory properties and potently reduce the cytokine-mediated IL-6 release [157]) against post-stroke depression was confirmed [158]. Thus, suppression of IL-6 activity could possibly lead to clinical recovery and may be considered as a novel pharmacotherapeutic option. Utilizing IL-6 receptor antibodies (for instance, Tocilizumab) or IL-6 antibodies (for instance, Sirukumab or Siltuximab) for reduction of IL-6 activity seems to be a novel strategy.
Blockade of IL-6 receptor by the humanized anti-IL-6 antibody, Tocilizumab has been used in treatment of rheumatoid arthritis (RA) [159163] and systemic juvenile idiopathic arthritis [164166]. Extensive clinical studies have established both short-term and long-term efficacy and safety of Tocilizumab [±conventional disease-modifying anti-rheumatic drugs (DMARDs)] in adults with moderate to severe RA. Additionally, Tocilizumab was shown to be effective as a monotherapy in patients with systemic juvenile idiopathic arthritis and also in patients whose disease has been refractory to other therapies [164]. Moreover, the safety profile of tocilizumab was reported to be consistent over time and also consistent with safety profile of other immunomodulatory agents [162]. It is also important to note that oral tocilizumab has been shown to inhibit experimental autoimmune encephalitis by elevating Th2 anti-inflammatory cytokines and decreasing pro-inflammatory Th1 cytokines [167]. With regard to crucial role of IL-6 in regulating metabolic homeostasis, side effects such as significant weight gain followed by hypertrygliceridemia and hypercholesterolemia may be observed in patients treated with tocilizumab [168]. Blockade of IL-6 trans-signaling, while classical IL-6R signaling stays intact seems to be crucial for the goal of maintaining gut mucosal integrity and epithelial regeneration [65]. Indeed, few randomized clinical trials were conducted on anti-depressant properties of tocilizumab. According to the results of a recent meta-analysis of anti-depressant activity of anti-cytokine therapies, treatment with tocilizumab showed statistically significant improvements in depressive symptoms [169].
Another promising human monoclonal antibody against Il-6, namely Sirukumab has been reported to be a safe and well-tolerated agent, capable of modulating the immune response in healthy populations as well as in patients with inflammatory disorders (e.g., rheumatoid arthritis). It targets the IL-6 signaling pathway by inhibition of both the pro- and anti-inflammatory effects of IL-6 [170]. Effects of Sirukumab on cytokine networks provide a well-founded rationale for its potential use in pharmacotherapy of psychiatric disorders promising possible advantages across varying domains of the biobehavioral research criteria [171]. In a phase 2, double-blind, placebo-controlled trial, the efficacy of Sirukumab and Siltuximab on depressive symptoms was studied in patients with rheumatoid arthritis or multicentric Castleman’s disease respectively. Compared with placebo, both IL-6 neutralizing antibodies were found to make significantly greater improvements on depressive symptoms in the patients [172]. Results of a recent mega-analysis of 18 randomized, placebo-controlled clinical trials of efficacy of immunomodulatory drugs on depressive symptoms in patients with inflammatory disorders demonstrated promising results (N = 10,743 participants). According to their findings, anti-IL-6 antibodies (sirukumab and siltuximab) had large and statistically significant effect sizes on core depressive symptoms before correction for physical health outcomes. Additionally, their effects remained significant in non-responders for the primary disease states evaluated [173]. Although further research is needed in this area, potential application of anti-IL-6 antibodies could possibly open new avenues in pharmacotherapy of MDD.

Possible role of IL-6 together with gut microbiota in pathogenesis of depression

The human intestine harbors nearly 100 trillion bacteria [174] consisting assemblages of microorganisms associated with various niches in and on the body with long-term implications to health [175]. Evidence is emerging regarding correlations of microbial activities with progressive structural and functional processes in the brain of both animal models and humans [175]. There is a large body of evidence for the role of gut microbiota composition in pathogenesis of depression [176180]. Moreover, there is growing body of literature for the influence of the gut microbiome on cytokine signaling [181, 182].
The dominant gut microbial phyla are known to be Firmicutes and Bacteroideteds [183, 184]. The Firmicutes/Bacteroideteds ratio is of great relevance in signaling human gut microbiota status [185]. For instance, increased levels of Firmicutes/Bacteroideteds ratio have been reported in patients suffering from irritable bowel syndrome (IBD) and seem to have some correlations with development of depression and anxiety [186, 187]. Additionally, Firmicutes/Bacteroideteds ratio is associated with overall alterations in bacterial profiles at different life stages [185]. In a novel study, researchers reported decreased Firmicutes/Bacteroideteds ratio in mice following social defeat stress; thus, proposing possible role of Firmicutes/Bacteroideteds in depressive-like behavior. Furthermore, administration of anti-mouse IL-6 receptor antibody (MR16-1) attenuated the decreased ratio of Firmicutes/Bacteroideteds in susceptible mice. Thus, the researchers concluded that anti-mouse IL-6 receptor antibody may have anti-depressant-like effects by normalizing the Firmicutes/Bacteroideteds ratio via modulation of the immune system [3].
Decreased number of Oscillospira was detected in patients with depression [180] which suggests for possible role of Oscillospira in pathogenesis of depression. Two animal studies yielded same results. A recent study investigated therapeutic effects of finasteride on depressive-like behavior in rats together with 1 month of treatment withdrawal. Withdrawal from finasteride was associated with increased depressive-like behavioral responses. Therapeutic use of finasteride was linked with elevations in the phylum Bacteroidetes and in the family Prevotellaceae, and withdrawal was found to be correlated with decreases in the family Ruminococcaceae and the genera Oscillospira and Lachnospira [188]. In another study, socially stressed mice developing depression-like symptoms showed increases at the genus level of fecal Oscillospira. Interestingly, IV administration of anti-mouse IL-6 receptor antibody (MR16-1) normalized depression-like behavior and resulted in significant decrease in Oscillospira levels towards pre-stressor levels [3]. Moreover, increased number of Sutterella was reported in fecal samples [189] and intestinal biopsy samples of children with Autism spectrum disorder [190]. Additionally, elevated number of Staphylococcus and Sutterella were found in mice following social defeat stress. It is likely that Staphylococcus and Sutterella play a role in the depressive-like behavior via infection-induced inflammation. Interestingly, administration of anti-mouse IL-6 receptor antibody resulted in attenuation of elevated number of Staphylococcus and Sutterella following social defeat stress in mice [3].
These findings advocate that peripheral IL-6 may have a significant role in pathogenesis of MDD and blockade of IL-6 receptor in the periphery may exhibit rapid-onset effects by attenuating the altered composition of gut microbiota. Taking into account the role of gut-microbiota in immunomodulation, it is highly probable that gut-microbiota-brain- axis plays a role in anti-depressant actions of treatment with anti-IL-6 receptor [3]. A brief overview of the role of IL-6 together with gut microbiota in pathogenesis of depression is shown in Fig. 2.

Elevated levels of IL-6 in patients with COVID-19 infection

The world-wide effect of the coronavirus disease 2019 (COVID-19) pandemic is enormous and is not solely limited to the increased mortality and morbidity rates, but also extends into the mental health of the global population [191]. Considerable amount of clinical data is emerging regarding the manifestation of depression in patients during [192194] and post-COVID 19 infection [192195]. It is estimated that about 48% of confirmed COVID-19 cases displayed overt psychological symptoms such as depression and often expressed feelings of regret, loneliness, helplessness, and irritation [196].
There is growing body of literature regarding dual role of IL-6 in COVID-19 infection and depression [192]. Normal plasma levels of IL-6 in adults range between 1 and 10 pg/ml; whereas in a systemic inflammation this amount increases to several ng/ml [197] and even higher concentrations were reported in COVID-19 patients [198]. Indeed, cytokine release syndrome (CRS) is common in COVID-19 patients and increased levels of serum IL-6 have been identified to be significantly associated with acute respiratory distress syndrome (ARDS), respiratory failure, and poor disease outcome in numerous studies [192, 199201]. Studies suggest that new onset depression is most probably caused by inflammation initiated during the active phase of the infection leading to a cytokine surge [202]. Indeed, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infects primarily human monocytes and dendritic cells causing dendritic cell dysfunction, leading to T cell apoptosis and exhaustion; thus, contributing to the immunopathology [203]. Alpert et al. (2020) described two cases of COVID-19 patients with elevated amounts of IL-6 (25 pg/mL and 26.7 pg/mL respectively) who were diagnosed with major depressive disorder (according to the Diagnostic and Statistical Manual of Mental Disorder, 5th Edition (DSM-5)) during COVID 19 infection. Both patients’ depressive symptoms subsided about 6 weeks after initiation of anti-depressant pharmacotherapy and normalization of the inflammatory cytokines [192]. The authors concluded that lower cytokine activity ameliorates depressive symptoms as normalization of IL-6 plasma levels decreased depression with or without anti-depressants [192]. Moreover, Benedett et al. (2020) studied effects of treatment with cytokine-blocking agents on the psychopathological status of the patients with COVID-19 infection. Their results were in favor of the protective effects of treatment with cytokine-blocking agents in early phases of COVID-19 against the later onset of depression [204].
It is indeed crucial to maintain a multidisciplinary approach in management of the psychological effects of this debilitating pandemic. Treatment strategies addressing the immunopathology of SARS-CoV-2 infection will be promising during the acute phase of the disease [192, 205]. Currently, there are few studies considering psychological and neuropsychiatric implications of COVID-19; however, it is very likely to expect an increased incidence of mental pathologies both during and post-COVID-19 infection.

Conclusion

Preclinical and clinical studies present strong evidence that inflammation is altered in a subset of patients with MDD and there is mounting body of literature for the role of pro-inflammatory cytokines namely IL-6 in pathophysiology of depression. Nevertheless, there still exists gap in our understanding of the mechanisms by which IL-6 signaling and its molecular components could possibly contribute to depression manifestation. A number of humanized monoclonal antibodies are undergoing clinical trials for potential pharmacotherapy of mood disorders. Biologics including IL-6 receptor antibodies or IL-6 antibodies are currently approved to treat inflammatory disorders such as RA and are undergoing clinical trials as a novel target for MDD treatment. However, these novel therapeutic targets may also raise the possibility of potential side effects. By investigating the interface of peripheral cytokines, namely IL-6 and brain cellular processes contributing to depression, one might be able to develop novel therapeutic options for treatment of mood disorders by sequestering and preventing this peripherally derived inflammatory marker from acting upon mood circuits in the CNS. In sum, therapeutic deficiency in treatment outcomes reflects the growing demand for revitalizing psychiatric therapeutics with novel options that could potentially open new avenues in treatment of this debilitating disorder and enhancement of patients’ quality of life.

Acknowledgements

Not applicable
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Depression and Other Common Mental Disorders: Global Health Estimates. 2017. Geneva: World Health Organization, Licence: CC BY-NC-SA 3.0 IGO. Depression and Other Common Mental Disorders: Global Health Estimates. 2017. Geneva: World Health Organization, Licence: CC BY-NC-SA 3.0 IGO.
2.
Zurück zum Zitat Liu Q, He H, Yang J, Feng X, Zhao F, Lyu J. Changes in the global burden of depression from 1990 to 2017: Findings from the Global Burden of Disease study. J Psychiatr Res. 2020;126:134–40.PubMedCrossRef Liu Q, He H, Yang J, Feng X, Zhao F, Lyu J. Changes in the global burden of depression from 1990 to 2017: Findings from the Global Burden of Disease study. J Psychiatr Res. 2020;126:134–40.PubMedCrossRef
3.
Zurück zum Zitat Zhang JC, Yao W, Dong C, Yang C, Ren Q, Ma M, Hashimoto K. Blockade of interleukin-6 receptor in the periphery promotes rapid and sustained antidepressant actions: a possible role of gut-microbiota-brain axis. Transl Psychiatry. 2017;7:e1138.PubMedPubMedCentralCrossRef Zhang JC, Yao W, Dong C, Yang C, Ren Q, Ma M, Hashimoto K. Blockade of interleukin-6 receptor in the periphery promotes rapid and sustained antidepressant actions: a possible role of gut-microbiota-brain axis. Transl Psychiatry. 2017;7:e1138.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Sim K, Lau WK, Sim J, Sum MY, Baldessarini RJ. Prevention of Relapse and Recurrence in Adults with Major Depressive Disorder: Systematic Review and Meta-Analyses of Controlled Trials. Int J Neuropsychopharmacol. 2016;19:pyv076. Sim K, Lau WK, Sim J, Sum MY, Baldessarini RJ. Prevention of Relapse and Recurrence in Adults with Major Depressive Disorder: Systematic Review and Meta-Analyses of Controlled Trials. Int J Neuropsychopharmacol. 2016;19:pyv076.
6.
Zurück zum Zitat Fonseka TM, McIntyre RS, Soczynska JK, Kennedy SH. Novel investigational drugs targeting IL-6 signaling for the treatment of depression. Expert Opin Investig Drugs. 2015;24:459–75.PubMedCrossRef Fonseka TM, McIntyre RS, Soczynska JK, Kennedy SH. Novel investigational drugs targeting IL-6 signaling for the treatment of depression. Expert Opin Investig Drugs. 2015;24:459–75.PubMedCrossRef
7.
Zurück zum Zitat Miller AH, Raison CL. The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nat Rev Immunol. 2016;16:22–34.PubMedPubMedCentralCrossRef Miller AH, Raison CL. The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nat Rev Immunol. 2016;16:22–34.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Majd M, Hashemian F, Hosseini SM, Vahdat Shariatpanahi M. Sharifi A. A randomized, double-blind, placebo-controlled trial of celecoxib augmentation of sertraline in treatment of drug-naive depressed women: a pilot study. Iran J Pharm Res. 2015;14:891–9.PubMedPubMedCentral Majd M, Hashemian F, Hosseini SM, Vahdat Shariatpanahi M. Sharifi A. A randomized, double-blind, placebo-controlled trial of celecoxib augmentation of sertraline in treatment of drug-naive depressed women: a pilot study. Iran J Pharm Res. 2015;14:891–9.PubMedPubMedCentral
9.
Zurück zum Zitat Wohleb ES, Franklin T, Iwata M, Duman RS. Integrating neuroimmune systems in the neurobiology of depression. Nat Rev Neurosci. 2016;17:497–511.PubMedCrossRef Wohleb ES, Franklin T, Iwata M, Duman RS. Integrating neuroimmune systems in the neurobiology of depression. Nat Rev Neurosci. 2016;17:497–511.PubMedCrossRef
10.
Zurück zum Zitat Zhao G, Liu X. Neuroimmune advance in depressive disorder. Adv Exp Med Biol. 2019;1180:85–98.PubMedCrossRef Zhao G, Liu X. Neuroimmune advance in depressive disorder. Adv Exp Med Biol. 2019;1180:85–98.PubMedCrossRef
12.
13.
Zurück zum Zitat Cassano T, Calcagnini S, Carbone A, Bukke VN, Orkisz S, Villani R, Romano A, Avolio C, Gaetani S. Pharmacological treatment of depression in Alzheimer’s disease: a challenging task. Front Pharmacol. 2019;10:1067.PubMedPubMedCentralCrossRef Cassano T, Calcagnini S, Carbone A, Bukke VN, Orkisz S, Villani R, Romano A, Avolio C, Gaetani S. Pharmacological treatment of depression in Alzheimer’s disease: a challenging task. Front Pharmacol. 2019;10:1067.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Hirschfield RM. History and evolution of the monoamine hypothesis of depression. J Clin Psychiatry. 2000;61:4–6. Hirschfield RM. History and evolution of the monoamine hypothesis of depression. J Clin Psychiatry. 2000;61:4–6.
15.
Zurück zum Zitat Massart R, Mongeau R, Lanfumey L. Beyond the monoaminergic hypothesis: neuroplasticity and epigenetic changes in a transgenic mouse model of depression. PhilosTrans R Soc Lond Ser B Biol Sci. 2012;367:2485–94.CrossRef Massart R, Mongeau R, Lanfumey L. Beyond the monoaminergic hypothesis: neuroplasticity and epigenetic changes in a transgenic mouse model of depression. PhilosTrans R Soc Lond Ser B Biol Sci. 2012;367:2485–94.CrossRef
16.
Zurück zum Zitat Boku S, Nakagawa S, Toda H, Hishimoto A. Neural basis of major depressive disorder: Beyond monoamine hypothesis. Psychiatry Clin Neurosci. 2018;72:3–12.PubMedCrossRef Boku S, Nakagawa S, Toda H, Hishimoto A. Neural basis of major depressive disorder: Beyond monoamine hypothesis. Psychiatry Clin Neurosci. 2018;72:3–12.PubMedCrossRef
17.
Zurück zum Zitat Kopschina Feltes P, Doorduin J, Klein HC, Juárez-Orozco LE, Dierckx RA. Moriguchi- Jeckel CM, de Vries EF. Anti-inflammatory treatment for major depressive disorder: implications for patients with an elevated immune profile and non-responders to standard antidepressant therapy. J Psychopharmacol. 2017;31:1149–65.PubMedPubMedCentralCrossRef Kopschina Feltes P, Doorduin J, Klein HC, Juárez-Orozco LE, Dierckx RA. Moriguchi- Jeckel CM, de Vries EF. Anti-inflammatory treatment for major depressive disorder: implications for patients with an elevated immune profile and non-responders to standard antidepressant therapy. J Psychopharmacol. 2017;31:1149–65.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Maes M, Van der Planken M, Stevens WJ, Peeters D, DeClerck LS, Bridts CH, Schotte C, Cosyns P. Leukocytosis, monocytosis and neutrophilia: hallmarks of severe depression. J Psychiatr Res. 1992;26:125–134. Maes M, Van der Planken M, Stevens WJ, Peeters D, DeClerck LS, Bridts CH, Schotte C, Cosyns P. Leukocytosis, monocytosis and neutrophilia: hallmarks of severe depression. J Psychiatr Res. 1992;26:125–134.
19.
Zurück zum Zitat Maes M, Lambrechts J, Bosmans E, Jacobs J, Suy E, Vandervorst C, de Jonckheere C, Minner B, Raus J. Evidence for a systemic immune activation during depression: results of leukocyte enumeration by flow cytometry in conjunction with monoclonal antibody staining. Psychol Med. 1992;22:45–53.PubMedCrossRef Maes M, Lambrechts J, Bosmans E, Jacobs J, Suy E, Vandervorst C, de Jonckheere C, Minner B, Raus J. Evidence for a systemic immune activation during depression: results of leukocyte enumeration by flow cytometry in conjunction with monoclonal antibody staining. Psychol Med. 1992;22:45–53.PubMedCrossRef
20.
Zurück zum Zitat Maes M, Scharpé S, Meltzer HY, Bosmans E, Suy E, Calabrese J, Cosyns P. Relationships between interleukin-6 activity, acute phase proteins, and function of the hypothalamic- pituitary-adrenal axis in severe depression. Psychiatry Res. 1993;49:11–27.PubMedCrossRef Maes M, Scharpé S, Meltzer HY, Bosmans E, Suy E, Calabrese J, Cosyns P. Relationships between interleukin-6 activity, acute phase proteins, and function of the hypothalamic- pituitary-adrenal axis in severe depression. Psychiatry Res. 1993;49:11–27.PubMedCrossRef
21.
Zurück zum Zitat Slavich GM, Irwin MR. From stress to inflammation and major depressive disorder: a social signal transduction theory of depression. Psychol Bull. 2014;140:774–815.PubMedPubMedCentralCrossRef Slavich GM, Irwin MR. From stress to inflammation and major depressive disorder: a social signal transduction theory of depression. Psychol Bull. 2014;140:774–815.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Irwin MR, Miller AH. Depressive disorders and immunity: 20 years of progress and discovery. Brain Behav Immun. 2007;21:374–83.PubMedCrossRef Irwin MR, Miller AH. Depressive disorders and immunity: 20 years of progress and discovery. Brain Behav Immun. 2007;21:374–83.PubMedCrossRef
23.
Zurück zum Zitat Henter ID, de Sousa RT, Gold PW, Brunon AR, Zarate CA, Machado-Vieira R. Mood therapeutics: novel pharmacological approaches for treating depression. Expert Rev Clin Pharmacol. 2017;10:153-166. Henter ID, de Sousa RT, Gold PW, Brunon AR, Zarate CA, Machado-Vieira R. Mood therapeutics: novel pharmacological approaches for treating depression. Expert Rev Clin Pharmacol. 2017;10:153-166.
24.
Zurück zum Zitat Himmerich H, Patsalos O, Lichtblau N, Ibrahim MAA, Dalton B. Cytokine research in depression: principles, challenges, and open questions. Front Psychiatry. 2019;7(10):30.CrossRef Himmerich H, Patsalos O, Lichtblau N, Ibrahim MAA, Dalton B. Cytokine research in depression: principles, challenges, and open questions. Front Psychiatry. 2019;7(10):30.CrossRef
25.
Zurück zum Zitat Zou W, Feng R, Yang Y. Changes in the serum levels of inflammatory cytokines in antidepressant drug-naïve patients with major depression. PLoS One. 2018;13:e0197267.PubMedPubMedCentralCrossRef Zou W, Feng R, Yang Y. Changes in the serum levels of inflammatory cytokines in antidepressant drug-naïve patients with major depression. PLoS One. 2018;13:e0197267.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Miller AH, Maletic V, Raison CL. Inflammation and its discontents: the role of cytokines in the pathophysiology of major depression. Biol Psychiatr. 2009;65:732–41.CrossRef Miller AH, Maletic V, Raison CL. Inflammation and its discontents: the role of cytokines in the pathophysiology of major depression. Biol Psychiatr. 2009;65:732–41.CrossRef
27.
Zurück zum Zitat Horowitz MA, Zunszain PA. Neuroimmune and neuroendocrine abnormalities in depression: two sides of the same coin. Ann N Y Acad Sci. 2015;1351:68–79.PubMedCrossRef Horowitz MA, Zunszain PA. Neuroimmune and neuroendocrine abnormalities in depression: two sides of the same coin. Ann N Y Acad Sci. 2015;1351:68–79.PubMedCrossRef
29.
Zurück zum Zitat Young JJ, Bruno D. Pomara N. A review of the relationship between proinflammatory cytokines and major depressive disorder. J Affect Disord. 2014;169:15–20.PubMedCrossRef Young JJ, Bruno D. Pomara N. A review of the relationship between proinflammatory cytokines and major depressive disorder. J Affect Disord. 2014;169:15–20.PubMedCrossRef
30.
Zurück zum Zitat Kim YK, Na KS, Shin KH, Jung HY, Choi SH, Kim JB. Cytokine imbalance in the pathophysiology of major depressive disorder. Prog Neuropsychopharmacol Biol Psychiatry. 2007;31:1044–53.PubMedCrossRef Kim YK, Na KS, Shin KH, Jung HY, Choi SH, Kim JB. Cytokine imbalance in the pathophysiology of major depressive disorder. Prog Neuropsychopharmacol Biol Psychiatry. 2007;31:1044–53.PubMedCrossRef
31.
Zurück zum Zitat Himmerich H, Kohls E, Hegerl U, Rummel-Kluge C. Prädiktive Faktoren der Depression und ihrer Therapie. Der Nervenarzt. 2014;85:1249–54.PubMedCrossRef Himmerich H, Kohls E, Hegerl U, Rummel-Kluge C. Prädiktive Faktoren der Depression und ihrer Therapie. Der Nervenarzt. 2014;85:1249–54.PubMedCrossRef
32.
Zurück zum Zitat Muñoz RF, Cuijpers P, Smit F, Barrera AZ, Leykin Y. Prevention of major depression. Ann Rev Clin Psychol. 2010;6:181–212.CrossRef Muñoz RF, Cuijpers P, Smit F, Barrera AZ, Leykin Y. Prevention of major depression. Ann Rev Clin Psychol. 2010;6:181–212.CrossRef
33.
Zurück zum Zitat Brebner K, Hayley S, Zacharko R, Merali Z, Anisman H. Synergistic effects of interleukin-1β, interleukin-6, and tumor necrosis factor-α: Central monoamine, corticosterone, and behavioral variations. Neuropsychopharmacology. 2000;22:566–80.PubMedCrossRef Brebner K, Hayley S, Zacharko R, Merali Z, Anisman H. Synergistic effects of interleukin-1β, interleukin-6, and tumor necrosis factor-α: Central monoamine, corticosterone, and behavioral variations. Neuropsychopharmacology. 2000;22:566–80.PubMedCrossRef
34.
Zurück zum Zitat Farooq RK, Asghar K, Kanwal S, Zulqernain A. Role of inflammatory cytokines in depression: focus on interleukin-1β. Biomed Rep. 2017;6:15–20.PubMedCrossRef Farooq RK, Asghar K, Kanwal S, Zulqernain A. Role of inflammatory cytokines in depression: focus on interleukin-1β. Biomed Rep. 2017;6:15–20.PubMedCrossRef
35.
Zurück zum Zitat Felger JC, Lotrich FE. Inflammatory cytokines in depression: neurobiological mechanisms and therapeutic implications. Neuroscience. 2013;246:199–229.PubMedCrossRef Felger JC, Lotrich FE. Inflammatory cytokines in depression: neurobiological mechanisms and therapeutic implications. Neuroscience. 2013;246:199–229.PubMedCrossRef
36.
Zurück zum Zitat Black C, Miller BJ. Meta-analysis of cytokines and chemokines in suicidality: distinguishing suicidal versus nonsuicidal patients. Biol Psychiatry. 2015;78:28–37.PubMedCrossRef Black C, Miller BJ. Meta-analysis of cytokines and chemokines in suicidality: distinguishing suicidal versus nonsuicidal patients. Biol Psychiatry. 2015;78:28–37.PubMedCrossRef
37.
Zurück zum Zitat Goldsmith DR, Rapaport MH. Miller BJ. A meta-analysis of blood cytokine network alterations in psychiatric patients: comparisons between schizophrenia, bipolar disorder and depression. Mol Psychiatry. 2016;21:1696–709.PubMedPubMedCentralCrossRef Goldsmith DR, Rapaport MH. Miller BJ. A meta-analysis of blood cytokine network alterations in psychiatric patients: comparisons between schizophrenia, bipolar disorder and depression. Mol Psychiatry. 2016;21:1696–709.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Dowlati Y, Herrmann N, Swardfager W, Liu H, Sham L, Reim EK. Lanctôt KL. A meta-analysis of cytokines in major depression. Biol Psychiatry. 2010;67:446–57.PubMedCrossRef Dowlati Y, Herrmann N, Swardfager W, Liu H, Sham L, Reim EK. Lanctôt KL. A meta-analysis of cytokines in major depression. Biol Psychiatry. 2010;67:446–57.PubMedCrossRef
39.
Zurück zum Zitat Strawbridge R, Arnone D, Danese A, Papadopoulos A, Herane Vives A, Cleare AJ. Inflammation and clinical response to treatment in depression: A meta-analysis. Eur Neuropsychopharmacol. 2015;25:1532–43.PubMedCrossRef Strawbridge R, Arnone D, Danese A, Papadopoulos A, Herane Vives A, Cleare AJ. Inflammation and clinical response to treatment in depression: A meta-analysis. Eur Neuropsychopharmacol. 2015;25:1532–43.PubMedCrossRef
41.
Zurück zum Zitat Nishuty NL, Khandoker MH, Karmoker JR, Ferdous S, Shahriar M, Qusar S, Islam S. Kadir MF, Islam R. Evaluation of serum interleukin-6 and C-reactive protein levels in drug-naïve major depressive disorder patient. cureus. 2019;11:e3868.PubMedPubMedCentral Nishuty NL, Khandoker MH, Karmoker JR, Ferdous S, Shahriar M, Qusar S, Islam S. Kadir MF, Islam R. Evaluation of serum interleukin-6 and C-reactive protein levels in drug-naïve major depressive disorder patient. cureus. 2019;11:e3868.PubMedPubMedCentral
42.
Zurück zum Zitat Ramesh G, MacLean AG, Philipp MT. Cytokines and chemokines at the crossroads of neuroinflammation, neurodegeneration. and neuropathic pain. Mediators Inflamm. 2013;2013:480739.PubMed Ramesh G, MacLean AG, Philipp MT. Cytokines and chemokines at the crossroads of neuroinflammation, neurodegeneration. and neuropathic pain. Mediators Inflamm. 2013;2013:480739.PubMed
43.
Zurück zum Zitat Arango Duque G, Descoteaux A. Macrophage cytokines: involvement in immunity and infectious diseases. Front Immunol. 2014;7(5):491. Arango Duque G, Descoteaux A. Macrophage cytokines: involvement in immunity and infectious diseases. Front Immunol. 2014;7(5):491.
44.
Zurück zum Zitat Galic MA, Riazi K, Pittman QJ. Cytokines and brain excitability. Front Neuroendocrinol. 2012;33:116–25.PubMedCrossRef Galic MA, Riazi K, Pittman QJ. Cytokines and brain excitability. Front Neuroendocrinol. 2012;33:116–25.PubMedCrossRef
45.
Zurück zum Zitat Wilson CJ, Finch CE, Cohen HJ. Cytokines and cognition--the case for a head-to-toe inflammatory paradigm. J Am Geriatr Soc. 2002;50:2041–56.PubMedCrossRef Wilson CJ, Finch CE, Cohen HJ. Cytokines and cognition--the case for a head-to-toe inflammatory paradigm. J Am Geriatr Soc. 2002;50:2041–56.PubMedCrossRef
46.
Zurück zum Zitat Hopkins SJ. Central nervous system recognition of peripheral inflammation: a neural. hormonal collaboration. Acta Biomed. 2007;78:231–47.PubMed Hopkins SJ. Central nervous system recognition of peripheral inflammation: a neural. hormonal collaboration. Acta Biomed. 2007;78:231–47.PubMed
48.
Zurück zum Zitat Caldwell AB, Cheng Z, Vargas JD, Birnbaum HA, Hoffmann A. Network dynamics determine the autocrine and paracrine signaling functions of TNF. Genes Dev. 2014;28:2120–33.PubMedPubMedCentralCrossRef Caldwell AB, Cheng Z, Vargas JD, Birnbaum HA, Hoffmann A. Network dynamics determine the autocrine and paracrine signaling functions of TNF. Genes Dev. 2014;28:2120–33.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Miller AH, Haroon E, Raison CL, Felger JC. Cytokine targets in the brain: impact on neurotransmitters and neurocircuits. Depress Anxiety. 2013;30:297–306.PubMedPubMedCentralCrossRef Miller AH, Haroon E, Raison CL, Felger JC. Cytokine targets in the brain: impact on neurotransmitters and neurocircuits. Depress Anxiety. 2013;30:297–306.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Perskidskii IV, Barshtein IA. Biological manifestations of the tumor necrosis factor effect and its role in the pathogenesis of various diseases. Arkh Patol. 1992;54:5–10.PubMed Perskidskii IV, Barshtein IA. Biological manifestations of the tumor necrosis factor effect and its role in the pathogenesis of various diseases. Arkh Patol. 1992;54:5–10.PubMed
51.
52.
Zurück zum Zitat Kicielinska J, Pajtasz-Piasecka E. The role of IL-10 in the modulation of the immune response in normal conditions and the tumor environment. Postepy Higieny Med Doświadczal. 2014;68:879–92.CrossRef Kicielinska J, Pajtasz-Piasecka E. The role of IL-10 in the modulation of the immune response in normal conditions and the tumor environment. Postepy Higieny Med Doświadczal. 2014;68:879–92.CrossRef
53.
Zurück zum Zitat Munk ME, Emoto M. Functions of T-cell subsets and cytokines in mycobacterial infections. Eur Respir J Suppl. 1995;20:668s–−675s.PubMed Munk ME, Emoto M. Functions of T-cell subsets and cytokines in mycobacterial infections. Eur Respir J Suppl. 1995;20:668s–−675s.PubMed
54.
Zurück zum Zitat Krumm B, Xiang Y, Deng J. Structural biology of the IL-1 superfamily: key cytokines in the regulation of immune and inflammatory responses. Protein Sci. 2014;23:526–38.PubMedPubMedCentralCrossRef Krumm B, Xiang Y, Deng J. Structural biology of the IL-1 superfamily: key cytokines in the regulation of immune and inflammatory responses. Protein Sci. 2014;23:526–38.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Tilg H, Peschel C. Interferon-alpha and its effects on the cytokine cascade: a pro- and anti-inflammatory cytokine. Leukem Lymphoma. 1996;23:55–60.CrossRef Tilg H, Peschel C. Interferon-alpha and its effects on the cytokine cascade: a pro- and anti-inflammatory cytokine. Leukem Lymphoma. 1996;23:55–60.CrossRef
56.
Zurück zum Zitat Klimpel GR. Soluble factor(s) from LPS-activated macrophages induce cytotoxic T cell differentiation from alloantigen-primed spleen cells. J Immunol. 1980;125:1243–9.PubMedCrossRef Klimpel GR. Soluble factor(s) from LPS-activated macrophages induce cytotoxic T cell differentiation from alloantigen-primed spleen cells. J Immunol. 1980;125:1243–9.PubMedCrossRef
57.
Zurück zum Zitat Spooren A, Kolmus K, Laureys G, Clinckers R, De Keyser J, Haegeman G, Gerlo S. Interleukin-6. a mental cytokine. Brain Res Rev. 2011;67:157–83.PubMedCrossRef Spooren A, Kolmus K, Laureys G, Clinckers R, De Keyser J, Haegeman G, Gerlo S. Interleukin-6. a mental cytokine. Brain Res Rev. 2011;67:157–83.PubMedCrossRef
58.
Zurück zum Zitat Yamasaki K, Taga T, Hirata Y, Yawata H, Kawanishi Y, Seed B, Taniguchi T, Hirano T, Kishimoto T. Cloning and expression of the human interleukin-6 (BSF-2/IFN beta 2) receptor. Science. 1988;241:825–8.PubMedCrossRef Yamasaki K, Taga T, Hirata Y, Yawata H, Kawanishi Y, Seed B, Taniguchi T, Hirano T, Kishimoto T. Cloning and expression of the human interleukin-6 (BSF-2/IFN beta 2) receptor. Science. 1988;241:825–8.PubMedCrossRef
59.
Zurück zum Zitat Hibi M, Murakami M, Saito M, Hirano T, Taga T, Kishimoto T. Molecular cloning and expression of an IL-6 signal transducer, gp130. Cell. 1990;63:1149–57.PubMedCrossRef Hibi M, Murakami M, Saito M, Hirano T, Taga T, Kishimoto T. Molecular cloning and expression of an IL-6 signal transducer, gp130. Cell. 1990;63:1149–57.PubMedCrossRef
61.
Zurück zum Zitat Schett G. Physiological effects of modulating the interleukin-6 axis. Rheumatology. 2018;57:ii43–ii50. Schett G. Physiological effects of modulating the interleukin-6 axis. Rheumatology. 2018;57:ii43–ii50.
62.
Zurück zum Zitat Mullberg J, Schooltink H, Stoyan T, Gunther M, Graeve L, Buse G, Mackiewicz A. Heinrich PC, Rose-John S. The soluble interleukin-6 receptor is generated by shedding. Eur J Immunol. 1993;23:473–80.PubMedCrossRef Mullberg J, Schooltink H, Stoyan T, Gunther M, Graeve L, Buse G, Mackiewicz A. Heinrich PC, Rose-John S. The soluble interleukin-6 receptor is generated by shedding. Eur J Immunol. 1993;23:473–80.PubMedCrossRef
63.
Zurück zum Zitat Wolf J, Rose-John S, Garbers C. Interleukin-6 and its receptors: a highly regulated and dynamic system. Cytokine. 2014;70:11–20.PubMedCrossRef Wolf J, Rose-John S, Garbers C. Interleukin-6 and its receptors: a highly regulated and dynamic system. Cytokine. 2014;70:11–20.PubMedCrossRef
64.
Zurück zum Zitat Scheller J, Chalaris A, Schmidt-Arras D, Rose-John S. The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim Biophys Acta. 2011;1813:878–88.PubMedCrossRef Scheller J, Chalaris A, Schmidt-Arras D, Rose-John S. The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim Biophys Acta. 2011;1813:878–88.PubMedCrossRef
65.
Zurück zum Zitat Hunter CA, Jones SA. IL-6 as a keystone cytokine in health and disease. Nat Immunol. 2015;16:448–57.PubMedCrossRef Hunter CA, Jones SA. IL-6 as a keystone cytokine in health and disease. Nat Immunol. 2015;16:448–57.PubMedCrossRef
66.
Zurück zum Zitat Hegde S, Pahne J, Smola-Hess S. Novel immunosuppressive properties of interleukin- 6 in dendritic cells: inhibition of NF-kB binding activity and CCR7 expression. FASEB J. 2004;18:1439–41.PubMedCrossRef Hegde S, Pahne J, Smola-Hess S. Novel immunosuppressive properties of interleukin- 6 in dendritic cells: inhibition of NF-kB binding activity and CCR7 expression. FASEB J. 2004;18:1439–41.PubMedCrossRef
67.
Zurück zum Zitat Kimura A, Kishimoto T. IL-6: regulator of Treg/Th17 balance. Eur J Immunol. 2010;40:1830–5.PubMedCrossRef Kimura A, Kishimoto T. IL-6: regulator of Treg/Th17 balance. Eur J Immunol. 2010;40:1830–5.PubMedCrossRef
68.
Zurück zum Zitat Mangan PR, Harrington LE, O’Quinn DB, Helms WS, Bullard DC, Elson CO, Hatton RD, Wahl SM, Schoeb TR, Weaver CT. Transforming growth factor-beta induces development of the T(H)17 lineage. Nature. 2006;441:231–4.PubMedCrossRef Mangan PR, Harrington LE, O’Quinn DB, Helms WS, Bullard DC, Elson CO, Hatton RD, Wahl SM, Schoeb TR, Weaver CT. Transforming growth factor-beta induces development of the T(H)17 lineage. Nature. 2006;441:231–4.PubMedCrossRef
69.
Zurück zum Zitat Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, Weiner HL, Kuchroo VK. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441:235–8.PubMedCrossRef Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, Weiner HL, Kuchroo VK. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441:235–8.PubMedCrossRef
70.
Zurück zum Zitat Iwakura Y, Ishigame H, Saijo S, Nakae S. Functional specialization of interleukin-17 family members. Immunity. 2011;34:149–62.PubMedCrossRef Iwakura Y, Ishigame H, Saijo S, Nakae S. Functional specialization of interleukin-17 family members. Immunity. 2011;34:149–62.PubMedCrossRef
71.
Zurück zum Zitat Dominitzki S, Fantini MC, Neufert C, Nikolaev A, Galle PR, Scheller J, Monteleone G, Rose-John S, Neurath MF, Becker C. Cutting edge: trans-signaling via the soluble IL-6R abrogates the induction of FoxP3 in naïve CD4+CD25 T cells. J Immunol. 2007;179:2041–5.PubMedCrossRef Dominitzki S, Fantini MC, Neufert C, Nikolaev A, Galle PR, Scheller J, Monteleone G, Rose-John S, Neurath MF, Becker C. Cutting edge: trans-signaling via the soluble IL-6R abrogates the induction of FoxP3 in naïve CD4+CD25 T cells. J Immunol. 2007;179:2041–5.PubMedCrossRef
72.
Zurück zum Zitat Milovan BM, Ivan J, Gordana R, Jelena P, Slavica MJ, Nebojsa A, Lukic ML. Interleukin-6 in schizophrenia—is there a therapeutic relevance? Front Psychiatry. 2017;8:221.CrossRef Milovan BM, Ivan J, Gordana R, Jelena P, Slavica MJ, Nebojsa A, Lukic ML. Interleukin-6 in schizophrenia—is there a therapeutic relevance? Front Psychiatry. 2017;8:221.CrossRef
73.
Zurück zum Zitat Milenkovic VM, Stanton EH, Nothdurfter C, Rupprecht R, Wetzel CH. The role of chemokines in the pathophysiology of major depressive disorder. Int J Mol Sci. 2019;20:2283.PubMedCentralCrossRef Milenkovic VM, Stanton EH, Nothdurfter C, Rupprecht R, Wetzel CH. The role of chemokines in the pathophysiology of major depressive disorder. Int J Mol Sci. 2019;20:2283.PubMedCentralCrossRef
75.
Zurück zum Zitat Le Thuc O, Blondeau N, Nahon JL, Rovère C. The complex contribution of chemokines to neuroinflammation: switching from beneficial to detrimental effects. Ann N Y Acad Sci. 2015;1351:127–40.PubMedCrossRef Le Thuc O, Blondeau N, Nahon JL, Rovère C. The complex contribution of chemokines to neuroinflammation: switching from beneficial to detrimental effects. Ann N Y Acad Sci. 2015;1351:127–40.PubMedCrossRef
76.
Zurück zum Zitat Deshmane SL, Kremlev S, Amini S, Sawaya BE. Monocyte chemoattractant protein-1 (MCP-1): an overview. J Interferon Cytokine Res. 2009;29:313–26.PubMedPubMedCentralCrossRef Deshmane SL, Kremlev S, Amini S, Sawaya BE. Monocyte chemoattractant protein-1 (MCP-1): an overview. J Interferon Cytokine Res. 2009;29:313–26.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Stuart MJ, Baune BT. Chemokines and chemokine receptors in mood disorders, schizophrenia, and cognitive impairment: a systematic review of biomarker studies. Neurosci Biobehav Rev. 2014;42:93–115.PubMedCrossRef Stuart MJ, Baune BT. Chemokines and chemokine receptors in mood disorders, schizophrenia, and cognitive impairment: a systematic review of biomarker studies. Neurosci Biobehav Rev. 2014;42:93–115.PubMedCrossRef
78.
Zurück zum Zitat Ślusarczyk J, Trojan E, Głombik K, Budziszewska B, Kubera M, Lasoń W, Popiołek- Barczyk K, Mika J, Wędzony K, Basta-Kaim A. Prenatal stress is a vulnerability factor for altered morphology and biological activity of microglia cells. Front Cell Neurosci. 2015;9:82.PubMedPubMedCentralCrossRef Ślusarczyk J, Trojan E, Głombik K, Budziszewska B, Kubera M, Lasoń W, Popiołek- Barczyk K, Mika J, Wędzony K, Basta-Kaim A. Prenatal stress is a vulnerability factor for altered morphology and biological activity of microglia cells. Front Cell Neurosci. 2015;9:82.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Trojan E, Ślusarczyk J, Chamera K, Kotarska K, Głombik K, Kubera M, Basta-Kaim A. The modulatory properties of chronic antidepressant drugs treatment on the brain chemokine – chemokine receptor network: a molecular study in an animal model of depression. Front Pharmacol. 2017;8:779.PubMedPubMedCentralCrossRef Trojan E, Ślusarczyk J, Chamera K, Kotarska K, Głombik K, Kubera M, Basta-Kaim A. The modulatory properties of chronic antidepressant drugs treatment on the brain chemokine – chemokine receptor network: a molecular study in an animal model of depression. Front Pharmacol. 2017;8:779.PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Simon NM, McNamara K, Chow CW, Maser RS, Papakostas GI, Pollack MH, Nierenberg AA, Fava M. Wong KK. A detailed examination of cytokine abnormalities in major depressive disorder. Eur Neuropsychopharmacol. 2008;18:230–3.PubMedCrossRef Simon NM, McNamara K, Chow CW, Maser RS, Papakostas GI, Pollack MH, Nierenberg AA, Fava M. Wong KK. A detailed examination of cytokine abnormalities in major depressive disorder. Eur Neuropsychopharmacol. 2008;18:230–3.PubMedCrossRef
81.
Zurück zum Zitat Piletz JE, Halaris A, Iqbal O, Hoppensteadt D, Fareed J, Zhu H, Sinacore J, Devane CL. Pro- inflammatory biomakers in depression: treatment with venlafaxine. World J Biol Psychiatry. 2009;10:313–23.PubMedCrossRef Piletz JE, Halaris A, Iqbal O, Hoppensteadt D, Fareed J, Zhu H, Sinacore J, Devane CL. Pro- inflammatory biomakers in depression: treatment with venlafaxine. World J Biol Psychiatry. 2009;10:313–23.PubMedCrossRef
82.
Zurück zum Zitat De la Peña FR, Cruz-Fuentes C, Palacios L, Girón-Pérez MI, Medina-Rivero E. Ponce- Regalado MD, Alvarez-Herrera S, Pérez-Sánchez G, Becerril-Villanueva E, Maldonado-García JL, Jiménez-Martínez MC, Pavón L. Serum levels of chemokines in adolescents with major depression treated with fluoxetine. World J Psychiatry. 2020;10:175–86.PubMedPubMedCentralCrossRef De la Peña FR, Cruz-Fuentes C, Palacios L, Girón-Pérez MI, Medina-Rivero E. Ponce- Regalado MD, Alvarez-Herrera S, Pérez-Sánchez G, Becerril-Villanueva E, Maldonado-García JL, Jiménez-Martínez MC, Pavón L. Serum levels of chemokines in adolescents with major depression treated with fluoxetine. World J Psychiatry. 2020;10:175–86.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Romero-Sanchiz P, Nogueira-Arjona R, Araos P, Serrano A, Barrios V, Argente J, Garcia- Marchena N, Lopez-Tellez A, Rodriguez-Moreno S, Mayoral F, et al. Variation in chemokines plasma concentrations in primary care depressed patients associated with Internet-based cognitive-behavioral therapy. Sci Rep. 2020;10:1078.PubMedPubMedCentralCrossRef Romero-Sanchiz P, Nogueira-Arjona R, Araos P, Serrano A, Barrios V, Argente J, Garcia- Marchena N, Lopez-Tellez A, Rodriguez-Moreno S, Mayoral F, et al. Variation in chemokines plasma concentrations in primary care depressed patients associated with Internet-based cognitive-behavioral therapy. Sci Rep. 2020;10:1078.PubMedPubMedCentralCrossRef
84.
Zurück zum Zitat Leighton SP, Nerurkar L, Krishnadas R, Johnman C, Graham GJ, Cavanagh J. Chemokines in depression in health and in inflammatory illness: a systematic review and meta- analysis. Mol Psychiatry. 2018;23:48–58.PubMedCrossRef Leighton SP, Nerurkar L, Krishnadas R, Johnman C, Graham GJ, Cavanagh J. Chemokines in depression in health and in inflammatory illness: a systematic review and meta- analysis. Mol Psychiatry. 2018;23:48–58.PubMedCrossRef
85.
Zurück zum Zitat Ślusarczyk J, Trojan E, Chwastek J, Głombik K, Basta-Kaim AA. Potential Contribution of Chemokine Network Dysfunction to the Depressive Disorders. Curr Neuropharmacol. 2016;14:705–20.PubMedPubMedCentralCrossRef Ślusarczyk J, Trojan E, Chwastek J, Głombik K, Basta-Kaim AA. Potential Contribution of Chemokine Network Dysfunction to the Depressive Disorders. Curr Neuropharmacol. 2016;14:705–20.PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat Sasayama D, Hattori K, Wakabayashi C, Teraishi T, Hori H, Ota M, Yoshida S, Arima K, Higuchi T, Amano N, et al. Increased cerebrospinal fluid interleukin-6 levels in patients with schizophrenia and those with major depressive disorder. J Psychiatr Res. 2013;47:401–6.PubMedCrossRef Sasayama D, Hattori K, Wakabayashi C, Teraishi T, Hori H, Ota M, Yoshida S, Arima K, Higuchi T, Amano N, et al. Increased cerebrospinal fluid interleukin-6 levels in patients with schizophrenia and those with major depressive disorder. J Psychiatr Res. 2013;47:401–6.PubMedCrossRef
87.
Zurück zum Zitat Boufidou F, Lambrinoudaki I, Argeitis J, Zervas IM, Pliatsika P, Leonardou AA. Petropoulos G, Hasiakos D, Papadias K, Nikolaou C. CSF and plasma cytokines at delivery and postpartum mood disturbances. J Affect Disord. 2009;115:287–92.PubMedCrossRef Boufidou F, Lambrinoudaki I, Argeitis J, Zervas IM, Pliatsika P, Leonardou AA. Petropoulos G, Hasiakos D, Papadias K, Nikolaou C. CSF and plasma cytokines at delivery and postpartum mood disturbances. J Affect Disord. 2009;115:287–92.PubMedCrossRef
88.
Zurück zum Zitat Schmidt FM, Weschenfelder J, Sander C, Minkwitz J, Thormann J, Chittka T, Mergl R, Kirkby KC, Faßhauer M, Stumvoll M, et al. Inflammatory cytokines in general and central obesity and modulating effects of physical activity. PLoS ONE. 2015;10:e0121971.PubMedPubMedCentralCrossRef Schmidt FM, Weschenfelder J, Sander C, Minkwitz J, Thormann J, Chittka T, Mergl R, Kirkby KC, Faßhauer M, Stumvoll M, et al. Inflammatory cytokines in general and central obesity and modulating effects of physical activity. PLoS ONE. 2015;10:e0121971.PubMedPubMedCentralCrossRef
89.
Zurück zum Zitat Bastard JP, Jardel C, Delattre J, Hainque B, Bruckert E, Oberlin F. Evidence for a link between adipose tissue interleukin-6 content and serum C-reactive protein concentrations in obese subjects. Circulation. 1999;99:2221–2.PubMedCrossRef Bastard JP, Jardel C, Delattre J, Hainque B, Bruckert E, Oberlin F. Evidence for a link between adipose tissue interleukin-6 content and serum C-reactive protein concentrations in obese subjects. Circulation. 1999;99:2221–2.PubMedCrossRef
90.
Zurück zum Zitat Wellen KE, Hotamisligil GS. Obesity-induced inflammatory changes in adipose tissue. J Clin Invest. 2003112:1785–8. Wellen KE, Hotamisligil GS. Obesity-induced inflammatory changes in adipose tissue. J Clin Invest. 2003112:1785–8.
91.
Zurück zum Zitat Sweat V, Starr V, Bruehl H, Arentoft A, Tirsi A, Javier E. Convit A. C-reactive protein is linked to lower cognitive performance in overweight and obese women. Inflammation. 2008;31:198–207.PubMedCrossRef Sweat V, Starr V, Bruehl H, Arentoft A, Tirsi A, Javier E. Convit A. C-reactive protein is linked to lower cognitive performance in overweight and obese women. Inflammation. 2008;31:198–207.PubMedCrossRef
92.
Zurück zum Zitat Capuron L, Su S, Miller AH, Bremner JD, Goldberg J, Vogt GJ, Maisano C, Jones L. Murrah NV, Vaccarino V. Depressive symptoms and metabolic syndrome: is inflammation the underlying link? Biol Psychiatry. 2008;64:896–900.PubMedPubMedCentralCrossRef Capuron L, Su S, Miller AH, Bremner JD, Goldberg J, Vogt GJ, Maisano C, Jones L. Murrah NV, Vaccarino V. Depressive symptoms and metabolic syndrome: is inflammation the underlying link? Biol Psychiatry. 2008;64:896–900.PubMedPubMedCentralCrossRef
93.
Zurück zum Zitat Hou R, Garner M, Holmes C, Osmond C, Teeling J, Lau L, Baldwin DS. Peripheral inflammatory cytokines and immune balance in Generalised Anxiety Disorder: Case- controlled study. Brain Behav Immun. 2017;62:212–8.PubMedPubMedCentralCrossRef Hou R, Garner M, Holmes C, Osmond C, Teeling J, Lau L, Baldwin DS. Peripheral inflammatory cytokines and immune balance in Generalised Anxiety Disorder: Case- controlled study. Brain Behav Immun. 2017;62:212–8.PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Costello H, Gould RL, Abrol E, Howard R. Systematic review and meta-analysis of the association between peripheral inflammatory cytokines and generalised anxiety disorder. BMJ Open. 2019;9:e027925.PubMedPubMedCentralCrossRef Costello H, Gould RL, Abrol E, Howard R. Systematic review and meta-analysis of the association between peripheral inflammatory cytokines and generalised anxiety disorder. BMJ Open. 2019;9:e027925.PubMedPubMedCentralCrossRef
95.
Zurück zum Zitat Vieira MMM, Ferreira TB, Pacheco PAF, Barros PO, Almeida CRM, Araújo-Lima CF, Silva-Filho RG, Hygino J, Andrade RM, Linhares UC, et al. Enhanced Th17 phenotype in individuals with generalized anxiety disorder. J Neuroimmunol. 2010;229:212–8.PubMedCrossRef Vieira MMM, Ferreira TB, Pacheco PAF, Barros PO, Almeida CRM, Araújo-Lima CF, Silva-Filho RG, Hygino J, Andrade RM, Linhares UC, et al. Enhanced Th17 phenotype in individuals with generalized anxiety disorder. J Neuroimmunol. 2010;229:212–8.PubMedCrossRef
96.
Zurück zum Zitat Gray SM, Bloch MH. Systematic review of proinflammatory cytokines in obsessive- compulsive disorder. Curr Psychiatr Rep. 2012;14:220–8.CrossRef Gray SM, Bloch MH. Systematic review of proinflammatory cytokines in obsessive- compulsive disorder. Curr Psychiatr Rep. 2012;14:220–8.CrossRef
97.
Zurück zum Zitat Hori H, Kim Y. Inflammation and post-traumatic stress disorder. PCN Psychiatry and Clinical Neurosciences. 2019;73:143–53.CrossRef Hori H, Kim Y. Inflammation and post-traumatic stress disorder. PCN Psychiatry and Clinical Neurosciences. 2019;73:143–53.CrossRef
98.
Zurück zum Zitat Renna ME, O’Toole MS, Spaeth PE, Lekander M, Mennin DS. The association between anxiety, traumatic stress, and obsessive-compulsive disorders and chronic inflammation: a systematic review and meta-analysis. Depress Anxiety. 2018;35:1081–94.PubMedCrossRef Renna ME, O’Toole MS, Spaeth PE, Lekander M, Mennin DS. The association between anxiety, traumatic stress, and obsessive-compulsive disorders and chronic inflammation: a systematic review and meta-analysis. Depress Anxiety. 2018;35:1081–94.PubMedCrossRef
99.
Zurück zum Zitat Irwin MR. Why sleep is important for health: a psychoneuroimmunology perspective. Annu Rev Psychol. 2015;66:143–72.PubMedCrossRef Irwin MR. Why sleep is important for health: a psychoneuroimmunology perspective. Annu Rev Psychol. 2015;66:143–72.PubMedCrossRef
100.
Zurück zum Zitat Köhler CA, Freitas TH, Stubbs B, Maes M, Solmi M, Veronese N, de Andrade NQ, Morris G, Fernandes BS, Brunoni AR, et al. Peripheral alterations in cytokine and chemokine levels after antidepressant drug treatment for major depressive disorder: systematic review and meta-analysis. Mol Neurobiol. 2018;55:4195–206.PubMed Köhler CA, Freitas TH, Stubbs B, Maes M, Solmi M, Veronese N, de Andrade NQ, Morris G, Fernandes BS, Brunoni AR, et al. Peripheral alterations in cytokine and chemokine levels after antidepressant drug treatment for major depressive disorder: systematic review and meta-analysis. Mol Neurobiol. 2018;55:4195–206.PubMed
101.
Zurück zum Zitat Himmerich H, Minkwitz J, Kirkby K. Weight gain and metabolic changes during treatment with antipsychotics and antidepressants. Endocr Metabol Immune Disorder Drug Targets. 2015;15:252–60.CrossRef Himmerich H, Minkwitz J, Kirkby K. Weight gain and metabolic changes during treatment with antipsychotics and antidepressants. Endocr Metabol Immune Disorder Drug Targets. 2015;15:252–60.CrossRef
102.
Zurück zum Zitat Himmerich H, Koethe D, Schuld A, Yassouridis A, Pollmacher T. Plasma levels of leptin and endogenous immune modulators during treatment with carbamazepine or lithium. Psychopharmacology. 2005;179:447–51.PubMedCrossRef Himmerich H, Koethe D, Schuld A, Yassouridis A, Pollmacher T. Plasma levels of leptin and endogenous immune modulators during treatment with carbamazepine or lithium. Psychopharmacology. 2005;179:447–51.PubMedCrossRef
103.
Zurück zum Zitat Haapakoski R, Mathieu J, Ebmeier KP, Alenius H, Kivimaki M. Cumulative meta- analysis of interleukins 6 and 1beta, tumour necrosis factor alpha and C-reactive protein in patients with major depressive disorder. Brain Behav Immun. 2015;49:206–15.PubMedPubMedCentralCrossRef Haapakoski R, Mathieu J, Ebmeier KP, Alenius H, Kivimaki M. Cumulative meta- analysis of interleukins 6 and 1beta, tumour necrosis factor alpha and C-reactive protein in patients with major depressive disorder. Brain Behav Immun. 2015;49:206–15.PubMedPubMedCentralCrossRef
104.
Zurück zum Zitat Khandaker GM, Pearson RM, Zammit S, Lewis G, Jones PB. Association of serum interleukin 6 and C-reactive protein in childhood with depression and psychosis in young adult life: a population-based longitudinal study. JAMA Psychiatry. 2014;71:1121–8.PubMedPubMedCentralCrossRef Khandaker GM, Pearson RM, Zammit S, Lewis G, Jones PB. Association of serum interleukin 6 and C-reactive protein in childhood with depression and psychosis in young adult life: a population-based longitudinal study. JAMA Psychiatry. 2014;71:1121–8.PubMedPubMedCentralCrossRef
105.
Zurück zum Zitat Lanquillon S, Krieg JC, Bening-Abu-Shach U, Vedder H. Cytokine production and treatment response in major depressive disorder. Neuropsychopharmacology. 2000;22:370–9.PubMedCrossRef Lanquillon S, Krieg JC, Bening-Abu-Shach U, Vedder H. Cytokine production and treatment response in major depressive disorder. Neuropsychopharmacology. 2000;22:370–9.PubMedCrossRef
106.
Zurück zum Zitat Mutlu O, Gumuslu E, Ulak G, Celikyurt IK, Kokturk S, Kir HM, Akar F, Erden F. Effects of fluoxetine, tianeptine and olanzapine on unpredictable chronic mild stress-induced depression-like behavior in mice. Life Sci. 2012;91:1252–62.PubMedCrossRef Mutlu O, Gumuslu E, Ulak G, Celikyurt IK, Kokturk S, Kir HM, Akar F, Erden F. Effects of fluoxetine, tianeptine and olanzapine on unpredictable chronic mild stress-induced depression-like behavior in mice. Life Sci. 2012;91:1252–62.PubMedCrossRef
107.
Zurück zum Zitat Pan Y, Zhang WY, Xia X, Kong LD. Effects of icariin on hypothalamic-pituitary- adrenal axis action and cytokine levels in stressed Sprague-Dawley rats. Biol Pharm Bull. 2006;29:2399–403.PubMedCrossRef Pan Y, Zhang WY, Xia X, Kong LD. Effects of icariin on hypothalamic-pituitary- adrenal axis action and cytokine levels in stressed Sprague-Dawley rats. Biol Pharm Bull. 2006;29:2399–403.PubMedCrossRef
108.
Zurück zum Zitat Himmerich H, Fischer J, Bauer K, Kirkby KC, Sack U, Krugel U. Stress-induced cytokine changes in rats. Eur Cytokine Network. 2013;24:97–103.CrossRef Himmerich H, Fischer J, Bauer K, Kirkby KC, Sack U, Krugel U. Stress-induced cytokine changes in rats. Eur Cytokine Network. 2013;24:97–103.CrossRef
109.
Zurück zum Zitat Farooq RK, Isingrini E, Tanti A, Le Guisquet AM, Arlicot N, Minier F, Leman S, Chalon S, Belzung C. Camus V. Is unpredictable chronic mild stress (UCMS) a reliable model to study depression-induced neuroinflammation? Behav Brain Res. 2012;231:130–7.PubMedCrossRef Farooq RK, Isingrini E, Tanti A, Le Guisquet AM, Arlicot N, Minier F, Leman S, Chalon S, Belzung C. Camus V. Is unpredictable chronic mild stress (UCMS) a reliable model to study depression-induced neuroinflammation? Behav Brain Res. 2012;231:130–7.PubMedCrossRef
110.
Zurück zum Zitat Monje FJ, Cabatic M, Divisch I, Kim EJ, Herkner KR, Binder BR, Pollak DD. Constant darkness induces IL-6-dependent depression-like behavior through the NF-kappaB signaling pathway. J Neurosci. 2011;31:9075–83.PubMedPubMedCentralCrossRef Monje FJ, Cabatic M, Divisch I, Kim EJ, Herkner KR, Binder BR, Pollak DD. Constant darkness induces IL-6-dependent depression-like behavior through the NF-kappaB signaling pathway. J Neurosci. 2011;31:9075–83.PubMedPubMedCentralCrossRef
112.
Zurück zum Zitat Hodes GE, Pfau ML, Leboeuf M, Golden SA, Christoffel DJ, Bregman D, Rebusi N, Heshmati M, Aleyasin H, Warren BL, et al. Individual differences in the peripheral immune system promote resilience versus susceptibility to social stress. Proc Natl Acad Sci U S A. 2014;111:16136–41.PubMedPubMedCentralCrossRef Hodes GE, Pfau ML, Leboeuf M, Golden SA, Christoffel DJ, Bregman D, Rebusi N, Heshmati M, Aleyasin H, Warren BL, et al. Individual differences in the peripheral immune system promote resilience versus susceptibility to social stress. Proc Natl Acad Sci U S A. 2014;111:16136–41.PubMedPubMedCentralCrossRef
113.
Zurück zum Zitat Krishnan V. Molecular adaptations underlying susceptibility and resistance to social defeat in brain reward regions. Cell. 2007;131:391–404.PubMedCrossRef Krishnan V. Molecular adaptations underlying susceptibility and resistance to social defeat in brain reward regions. Cell. 2007;131:391–404.PubMedCrossRef
114.
Zurück zum Zitat Yang C, Shirayama Y, Zhang JC, Ren Q, Hashimoto K. Peripheral interleukin-6 promotes resilience versus susceptibility to inescapable electric stress. Acta Neuropsychiatr. 2015;27:312–6.PubMedCrossRef Yang C, Shirayama Y, Zhang JC, Ren Q, Hashimoto K. Peripheral interleukin-6 promotes resilience versus susceptibility to inescapable electric stress. Acta Neuropsychiatr. 2015;27:312–6.PubMedCrossRef
115.
Zurück zum Zitat Wang M, Wei J, Yang X, Ni P, Wang Y, Zhao L, Deng W, Guo W, Wang Q, Li T, et al. The level of IL-6 was associated with sleep disturbances in patients with major depressive disorder. Neuropsychiatric Disease and Treatment. 2019;15:1695–700.PubMedPubMedCentralCrossRef Wang M, Wei J, Yang X, Ni P, Wang Y, Zhao L, Deng W, Guo W, Wang Q, Li T, et al. The level of IL-6 was associated with sleep disturbances in patients with major depressive disorder. Neuropsychiatric Disease and Treatment. 2019;15:1695–700.PubMedPubMedCentralCrossRef
116.
Zurück zum Zitat Howren MB, Lamkin DM, Suls J. Associations of depression with C-reactive protein, IL-1, and IL-6: a meta-analysis. Psychosom Med. 2009;71:171–86.PubMedCrossRef Howren MB, Lamkin DM, Suls J. Associations of depression with C-reactive protein, IL-1, and IL-6: a meta-analysis. Psychosom Med. 2009;71:171–86.PubMedCrossRef
117.
Zurück zum Zitat 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;139:230–9.PubMedCrossRef 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;139:230–9.PubMedCrossRef
118.
Zurück zum Zitat Chocano-Bedoya PO, Mirzaei F, O’Reilly EJ, Lucas M, Okereke OI, Hu FB, Rimm EB, Ascherio A. C-reactive protein, interleukin-6, soluble tumor necrosis factor alpha receptor 2 and incident clinical depression. J Affect Disord. 2014;163:25–32.PubMedPubMedCentralCrossRef Chocano-Bedoya PO, Mirzaei F, O’Reilly EJ, Lucas M, Okereke OI, Hu FB, Rimm EB, Ascherio A. C-reactive protein, interleukin-6, soluble tumor necrosis factor alpha receptor 2 and incident clinical depression. J Affect Disord. 2014;163:25–32.PubMedPubMedCentralCrossRef
119.
Zurück zum Zitat Rudolf S, Greggersen W, Kahl KG, Huppe M, Schweiger U. Elevated il-6 levels in patients with atypical depression but not in patients with typical depression. Psychiatry Res. 2014;217:34–8.PubMedCrossRef Rudolf S, Greggersen W, Kahl KG, Huppe M, Schweiger U. Elevated il-6 levels in patients with atypical depression but not in patients with typical depression. Psychiatry Res. 2014;217:34–8.PubMedCrossRef
120.
Zurück zum Zitat Rush G, O’Donovan A, Nagle L, Conway C, McCrohan A, O’Farrelly C, Lucey JV, Malone KM. Alteration of immune markers in a group of melancholic depressed patients and their response to electroconvulsive therapy. J Affect Disord. 2016;205:60–8.PubMedPubMedCentralCrossRef Rush G, O’Donovan A, Nagle L, Conway C, McCrohan A, O’Farrelly C, Lucey JV, Malone KM. Alteration of immune markers in a group of melancholic depressed patients and their response to electroconvulsive therapy. J Affect Disord. 2016;205:60–8.PubMedPubMedCentralCrossRef
121.
Zurück zum Zitat Yang C, Tiemessen KM, Bosker FJ, Wardenaar KJ, Lie J, Schoevers RA. Interleukin, tumor necrosis factor-alpha and c-reactive protein profiles in melancholic and non-melancholic depression: A systematic review. J Psychosom Res. 2018;111:58–68.PubMedCrossRef Yang C, Tiemessen KM, Bosker FJ, Wardenaar KJ, Lie J, Schoevers RA. Interleukin, tumor necrosis factor-alpha and c-reactive protein profiles in melancholic and non-melancholic depression: A systematic review. J Psychosom Res. 2018;111:58–68.PubMedCrossRef
122.
Zurück zum Zitat Maes M, Bosmans E, De Jongh R, Kenis G, Vandoolaeghe E, Neels H. Increased serum IL- 6 and IL-1 receptor antagonist concentrations in major depression and treatment resistant depression. Cytokine. 1997;9:853–8.PubMedCrossRef Maes M, Bosmans E, De Jongh R, Kenis G, Vandoolaeghe E, Neels H. Increased serum IL- 6 and IL-1 receptor antagonist concentrations in major depression and treatment resistant depression. Cytokine. 1997;9:853–8.PubMedCrossRef
123.
Zurück zum Zitat Lichtblau N, Schmidt FM, Schumann R, Kirkby KC, Himmerich H. Cytokines as biomarkers in depressive disorder: current standing and prospects. Int Rev Psychiatry. 2013;25:592–603.PubMedCrossRef Lichtblau N, Schmidt FM, Schumann R, Kirkby KC, Himmerich H. Cytokines as biomarkers in depressive disorder: current standing and prospects. Int Rev Psychiatry. 2013;25:592–603.PubMedCrossRef
125.
Zurück zum Zitat Albert PR, Benkelfat C, Descarries L. The neurobiology of depression—revisiting the serotonin hypothesis. I. Cellular and molecular mechanisms. Philos Trans R Soc Lond B Biol Sci. 2012;367:2378–81.PubMedPubMedCentralCrossRef Albert PR, Benkelfat C, Descarries L. The neurobiology of depression—revisiting the serotonin hypothesis. I. Cellular and molecular mechanisms. Philos Trans R Soc Lond B Biol Sci. 2012;367:2378–81.PubMedPubMedCentralCrossRef
126.
Zurück zum Zitat Zhu CB, Carneiro AM, Dostmann WR, Hewlett WA. Blakely RD. p38 MAPK Activation Elevates Serotonin Transport Activity via a Trafficking-independent, Protein Phosphatase 2A-dependent Proces. The Journal of Biological Chemistry. 2005;280:15649–58.PubMedCrossRef Zhu CB, Carneiro AM, Dostmann WR, Hewlett WA. Blakely RD. p38 MAPK Activation Elevates Serotonin Transport Activity via a Trafficking-independent, Protein Phosphatase 2A-dependent Proces. The Journal of Biological Chemistry. 2005;280:15649–58.PubMedCrossRef
127.
Zurück zum Zitat Zhu CB, Blakely RD, Hewlett WA. The proinflammatory cytokines interleukin-1beta and tumor necrosis factor-alpha activate serotonin transporters. Neuropsychopharmacology. 2006;31:2121–31.PubMedCrossRef Zhu CB, Blakely RD, Hewlett WA. The proinflammatory cytokines interleukin-1beta and tumor necrosis factor-alpha activate serotonin transporters. Neuropsychopharmacology. 2006;31:2121–31.PubMedCrossRef
128.
Zurück zum Zitat Kong E, Sucic S, Monje FJ, Reisinger SN, Savalli G, Diao W, Khan D, Ronovsky M, Cabatic M, Koban F, et al. STAT3 controls IL6-dependent regulation of serotonin transporter function and depression-like behavior. Sci Rep. 2015;5:9009.PubMedPubMedCentralCrossRef Kong E, Sucic S, Monje FJ, Reisinger SN, Savalli G, Diao W, Khan D, Ronovsky M, Cabatic M, Koban F, et al. STAT3 controls IL6-dependent regulation of serotonin transporter function and depression-like behavior. Sci Rep. 2015;5:9009.PubMedPubMedCentralCrossRef
129.
Zurück zum Zitat Baudry A, Pietri M, Launay JM, Kellermann O, Schneider B. Multifaceted regulations of the serotonin transporter: impact on antidepressant response. Front Neurosci. 2019;13:91.PubMedPubMedCentralCrossRef Baudry A, Pietri M, Launay JM, Kellermann O, Schneider B. Multifaceted regulations of the serotonin transporter: impact on antidepressant response. Front Neurosci. 2019;13:91.PubMedPubMedCentralCrossRef
130.
Zurück zum Zitat Zalcman S, Green-Johnson JM, Murray L, Nance DM, Dyck D, Anisman H, Greenberg AH. Cytokine-specific central monoamine alterations induced by interleukin-1, -2 and – 6. Brain Res. 1994;643:40–9. Zalcman S, Green-Johnson JM, Murray L, Nance DM, Dyck D, Anisman H, Greenberg AH. Cytokine-specific central monoamine alterations induced by interleukin-1, -2 and – 6. Brain Res. 1994;643:40–9.
131.
Zurück zum Zitat Day JS, O’Neill E, Cawley C, Aretz NK, Kilroy D, Gibney SM, Harkin A, Conor TJ. Noradrenaline acting on astrocyticβ2-adrenoceptors induces neurite outgrowth in primary cortical neurons. Neuropharmacology. 2014;77:234–48.PubMedCrossRef Day JS, O’Neill E, Cawley C, Aretz NK, Kilroy D, Gibney SM, Harkin A, Conor TJ. Noradrenaline acting on astrocyticβ2-adrenoceptors induces neurite outgrowth in primary cortical neurons. Neuropharmacology. 2014;77:234–48.PubMedCrossRef
132.
Zurück zum Zitat Müller N, Myint AM, Krause D, Weidinger E, Schwarz MJ. Anti-inflammatory treatment in schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry. 2013;42:146–53.PubMedCrossRef Müller N, Myint AM, Krause D, Weidinger E, Schwarz MJ. Anti-inflammatory treatment in schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry. 2013;42:146–53.PubMedCrossRef
133.
Zurück zum Zitat Monje ML, Toda H, Palmer TD. Inflammatory blockade restores adult hippocampal neurogenesis. Science. 2003;302:1760–5.PubMedCrossRef Monje ML, Toda H, Palmer TD. Inflammatory blockade restores adult hippocampal neurogenesis. Science. 2003;302:1760–5.PubMedCrossRef
134.
Zurück zum Zitat Khairova RA, Machado-Vieira R, Du J. Manji HK. A potential role for pro-inflammatory cytokines in regulating synaptic plasticity in major depressive disorder. Int J Neuropsychopharmacol. 2009;12:561–78.PubMedCrossRef Khairova RA, Machado-Vieira R, Du J. Manji HK. A potential role for pro-inflammatory cytokines in regulating synaptic plasticity in major depressive disorder. Int J Neuropsychopharmacol. 2009;12:561–78.PubMedCrossRef
135.
Zurück zum Zitat Alexander GE, Crutcher MD, DeLong MR. Basal ganglia-thalamocortical circuits: parallel substrates for motor, oculomotor, “prefrontal” and “limbic” functions. Progress Brain Res. 1991;85:119–46.CrossRef Alexander GE, Crutcher MD, DeLong MR. Basal ganglia-thalamocortical circuits: parallel substrates for motor, oculomotor, “prefrontal” and “limbic” functions. Progress Brain Res. 1991;85:119–46.CrossRef
136.
Zurück zum Zitat Eisenberger NI, Lieberman MD. Why rejection hurts: a common neural alarm system for physical and social pain. Trends Cogn Sci. 2004;8:294–300.PubMedCrossRef Eisenberger NI, Lieberman MD. Why rejection hurts: a common neural alarm system for physical and social pain. Trends Cogn Sci. 2004;8:294–300.PubMedCrossRef
137.
Zurück zum Zitat Rothaug M, Becker-Pauly C, Rose-John S. The role of interleukin-6 signaling in nervous tissue. Biochim Biophys Acta. 2016;1863:1218–27.PubMedCrossRef Rothaug M, Becker-Pauly C, Rose-John S. The role of interleukin-6 signaling in nervous tissue. Biochim Biophys Acta. 2016;1863:1218–27.PubMedCrossRef
138.
Zurück zum Zitat Campbell IL, Abraham CR, Masliah E, Kemper P, Inglis JD, Oldstone MB, Mucke L. Neurologic disease induced in transgenic mice by cerebral overexpression of interleukin 6. Proc Natl Acad Sci U S A. 1993;90:10061–5.PubMedPubMedCentralCrossRef Campbell IL, Abraham CR, Masliah E, Kemper P, Inglis JD, Oldstone MB, Mucke L. Neurologic disease induced in transgenic mice by cerebral overexpression of interleukin 6. Proc Natl Acad Sci U S A. 1993;90:10061–5.PubMedPubMedCentralCrossRef
139.
Zurück zum Zitat Setiawan E, Attwells S, Wilson AA, Mizrahi R, Rusjan PM, Miler L, Xu C, Sharma S, Kish S, Houle S, et al. Association of translocator protein total distribution volume with duration of untreated major depressive disorder: a cross-sectional study. The Lancet Psychiatry. 2018;5:339–47.PubMedCrossRef Setiawan E, Attwells S, Wilson AA, Mizrahi R, Rusjan PM, Miler L, Xu C, Sharma S, Kish S, Houle S, et al. Association of translocator protein total distribution volume with duration of untreated major depressive disorder: a cross-sectional study. The Lancet Psychiatry. 2018;5:339–47.PubMedCrossRef
140.
Zurück zum Zitat Setiawan E, Wilson AA, Mizrahi R, Rusjan PM, Miler L, Rajkowska G, Suridjan I, Kennedy JL, Rekkas PV, Houle S, et al. Role of translocator protein density, a marker of neuroinflammation, in the brain during major depressive episodes. JAMA Psychiatry. 2015;72:268–75.PubMedPubMedCentralCrossRef Setiawan E, Wilson AA, Mizrahi R, Rusjan PM, Miler L, Rajkowska G, Suridjan I, Kennedy JL, Rekkas PV, Houle S, et al. Role of translocator protein density, a marker of neuroinflammation, in the brain during major depressive episodes. JAMA Psychiatry. 2015;72:268–75.PubMedPubMedCentralCrossRef
141.
Zurück zum Zitat Holmes SE, Hinz R, Conen S, Gregory CJ, Matthews JC, Anton-Rodriguez JM, Gerhard A, Talbot PS. Elevated translocator protein in anterior cingulate in major depression and a role for inflammation in suicidal thinking: a positron emission tomography study. Biol Psychiatry. 2018;83:61–9.PubMedCrossRef Holmes SE, Hinz R, Conen S, Gregory CJ, Matthews JC, Anton-Rodriguez JM, Gerhard A, Talbot PS. Elevated translocator protein in anterior cingulate in major depression and a role for inflammation in suicidal thinking: a positron emission tomography study. Biol Psychiatry. 2018;83:61–9.PubMedCrossRef
142.
Zurück zum Zitat Richards EM, Zanotti-Fregonara P, Fujita M, Newman L, Farmer C, Ballard ED, Machado- Vieira R, Yuan P, Niciu MJ, Lyoo CH, et al. PET radioligand binding to translocator protein (TSPO) is increased in unmedicated depressed subjects. EJNMMI Res. 2018;8:57.PubMedPubMedCentralCrossRef Richards EM, Zanotti-Fregonara P, Fujita M, Newman L, Farmer C, Ballard ED, Machado- Vieira R, Yuan P, Niciu MJ, Lyoo CH, et al. PET radioligand binding to translocator protein (TSPO) is increased in unmedicated depressed subjects. EJNMMI Res. 2018;8:57.PubMedPubMedCentralCrossRef
143.
Zurück zum Zitat Li H, Sagar AP, Kéri S. Translocator protein (18kDa TSPO) binding, a marker of microglia, is reduced in major depression during cognitive-behavioral therapy. Prog Neuropsychopharmacol Biol Psychiatry. 2018;83:1–7.PubMedCrossRef Li H, Sagar AP, Kéri S. Translocator protein (18kDa TSPO) binding, a marker of microglia, is reduced in major depression during cognitive-behavioral therapy. Prog Neuropsychopharmacol Biol Psychiatry. 2018;83:1–7.PubMedCrossRef
144.
Zurück zum Zitat Kakeda S, Watanabe K, Katsuki A, Sugimoto K, Igata N, Ueda I, Igata R, Abe O, Yoshimura R, Korogi Y. Relationship between interleukin (IL)-6 and brain morphology in drug-naïve, first-episode major depressive disorder using surface-based morphometry. Sci Rep. 2018;8:10054.PubMedPubMedCentralCrossRef Kakeda S, Watanabe K, Katsuki A, Sugimoto K, Igata N, Ueda I, Igata R, Abe O, Yoshimura R, Korogi Y. Relationship between interleukin (IL)-6 and brain morphology in drug-naïve, first-episode major depressive disorder using surface-based morphometry. Sci Rep. 2018;8:10054.PubMedPubMedCentralCrossRef
145.
Zurück zum Zitat Frodl T, Carballedo A, Hughes MM, Saleh K, Fagan A, Skokauskas N, McLoughlin DM, Meaney J, O'Keane V, Connor TJ. Reduced expression of glucocorticoid-inducible genes GILZ and SGK-1: high IL-6 levels are associated with reduced hippocampal volumes in major depressive disorder. Transl Psychiatry. 2012;2:e88.PubMedPubMedCentralCrossRef Frodl T, Carballedo A, Hughes MM, Saleh K, Fagan A, Skokauskas N, McLoughlin DM, Meaney J, O'Keane V, Connor TJ. Reduced expression of glucocorticoid-inducible genes GILZ and SGK-1: high IL-6 levels are associated with reduced hippocampal volumes in major depressive disorder. Transl Psychiatry. 2012;2:e88.PubMedPubMedCentralCrossRef
146.
Zurück zum Zitat Campbell S, Macqueen G. The role of the hippocampus in the pathophysiology of major depression. J Psychiatry Neurosci. 2004;29:417–26.PubMedPubMedCentral Campbell S, Macqueen G. The role of the hippocampus in the pathophysiology of major depression. J Psychiatry Neurosci. 2004;29:417–26.PubMedPubMedCentral
147.
Zurück zum Zitat MacQueen GM, Campbell S, McEwen BS, Macdonald K, Amano S, Joffe RT, Nahmias C, Young LT. Course of illness, hippocampal function, and hippocampal volume in major depression. Proc Natl Acad Sci U S A. 2003;100:1387-1392. MacQueen GM, Campbell S, McEwen BS, Macdonald K, Amano S, Joffe RT, Nahmias C, Young LT. Course of illness, hippocampal function, and hippocampal volume in major depression. Proc Natl Acad Sci U S A. 2003;100:1387-1392.
148.
Zurück zum Zitat Frodl T, Meisenzahl EM, Zetzsche T. Born C, Groll C, Jäger M, Leinsinger G, Bottlender R, Hahn K, Möller HJ. Hippocampal changes in patients with a first episode of major Depression. Am J Psychiatry. 2002;159:1112–8.PubMedCrossRef Frodl T, Meisenzahl EM, Zetzsche T. Born C, Groll C, Jäger M, Leinsinger G, Bottlender R, Hahn K, Möller HJ. Hippocampal changes in patients with a first episode of major Depression. Am J Psychiatry. 2002;159:1112–8.PubMedCrossRef
149.
Zurück zum Zitat Rajkowska G. Postmortem studies in mood disorders indicate altered numbers of neurons and glial cells. Biol Psychiatry. 2000;48:766–7.PubMedCrossRef Rajkowska G. Postmortem studies in mood disorders indicate altered numbers of neurons and glial cells. Biol Psychiatry. 2000;48:766–7.PubMedCrossRef
150.
Zurück zum Zitat Cameron HA, McKay RD. Restoring production of hippocampal neurons in old age. Nat Neurosci. 1999;2:894–7.PubMedCrossRef Cameron HA, McKay RD. Restoring production of hippocampal neurons in old age. Nat Neurosci. 1999;2:894–7.PubMedCrossRef
151.
Zurück zum Zitat Young EA, Haskett RF, Grunhaus L, Pande A, Weinberg VM, Watson SJ, Akil H. Increased evening activation of the hypothalamic-pituitary-adrenal axis in depressed patients. Arch Gen Psychiatry. 1994;51:701–7.PubMedCrossRef Young EA, Haskett RF, Grunhaus L, Pande A, Weinberg VM, Watson SJ, Akil H. Increased evening activation of the hypothalamic-pituitary-adrenal axis in depressed patients. Arch Gen Psychiatry. 1994;51:701–7.PubMedCrossRef
152.
Zurück zum Zitat Sapolsky RM. The possibility of neurotoxicity in the hippocampus in major depression: a primer on neuron death. Biol Psychiatry. 2000 Oct 15;48(8):755–65.PubMedCrossRef Sapolsky RM. The possibility of neurotoxicity in the hippocampus in major depression: a primer on neuron death. Biol Psychiatry. 2000 Oct 15;48(8):755–65.PubMedCrossRef
153.
Zurück zum Zitat McEwen BS. The neurobiology of stress: from serendipity to clinical relevance. Brain Res. 2000;886:172–89.PubMedCrossRef McEwen BS. The neurobiology of stress: from serendipity to clinical relevance. Brain Res. 2000;886:172–89.PubMedCrossRef
154.
Zurück zum Zitat Yang C, Hashimoto K. Peripheral IL-6 signaling: a promising therapeutic target for depression? Expert Opin Investig Drugs. 2015;24:989–90.PubMedCrossRef Yang C, Hashimoto K. Peripheral IL-6 signaling: a promising therapeutic target for depression? Expert Opin Investig Drugs. 2015;24:989–90.PubMedCrossRef
155.
Zurück zum Zitat Chourbaji S, Urani A, Inta I, Sanchis-Segura C, Brandwein C, Zink M, Schwaninger M, Gass P. IL-6 knockout mice exhibit resistance to stress-induced development of depression-like behaviors. Neurobiol Dis. 2006;23:587–94.PubMedCrossRef Chourbaji S, Urani A, Inta I, Sanchis-Segura C, Brandwein C, Zink M, Schwaninger M, Gass P. IL-6 knockout mice exhibit resistance to stress-induced development of depression-like behaviors. Neurobiol Dis. 2006;23:587–94.PubMedCrossRef
157.
Zurück zum Zitat Loppnow H, Zhang L, Buerke M, Lautenschläger M, Chen L, Frister A, Schlitt A, Luther T, Song N, Hofmann B, et al. Statins potently reduce the cytokine-mediated IL-6 release in SMC/MNC cocultures. J Cell Mol Med. 2011;15:994–1004.PubMedCrossRef Loppnow H, Zhang L, Buerke M, Lautenschläger M, Chen L, Frister A, Schlitt A, Luther T, Song N, Hofmann B, et al. Statins potently reduce the cytokine-mediated IL-6 release in SMC/MNC cocultures. J Cell Mol Med. 2011;15:994–1004.PubMedCrossRef
158.
Zurück zum Zitat Kang HJ, Bae KY, Kim SW, Kim JT, Park MS, Cho KH, Kim JM. Effects of interleukin-6, interleukin-18, and statin use, evaluated at acute stroke, on post-stroke depression during 1- year follow-up. Psychoneuroendocrinology. 2016;72:156–60.PubMedCrossRef Kang HJ, Bae KY, Kim SW, Kim JT, Park MS, Cho KH, Kim JM. Effects of interleukin-6, interleukin-18, and statin use, evaluated at acute stroke, on post-stroke depression during 1- year follow-up. Psychoneuroendocrinology. 2016;72:156–60.PubMedCrossRef
159.
Zurück zum Zitat Burmester GR, Rigby WF, van Vollenhoven RF, Kay J, Rubbert-Roth A, Kelman A, Dimonaco S, Mitchell N. Tocilizumab in early progressive rheumatoid arthritis: FUNCTION, a randomised controlled trial. Ann Rheum Di. 2016;75:1081-1091. Burmester GR, Rigby WF, van Vollenhoven RF, Kay J, Rubbert-Roth A, Kelman A, Dimonaco S, Mitchell N. Tocilizumab in early progressive rheumatoid arthritis: FUNCTION, a randomised controlled trial. Ann Rheum Di. 2016;75:1081-1091.
160.
Zurück zum Zitat Yazici Y, Curtis JR, Ince A, Baraf H, Malamet RL, Teng LL, Kavanaugh A. Efficacy of tocilizumab in patients with moderate to severe active rheumatoid arthritis and a previous inadequate response to disease-modifying antirheumatic drugs: the ROSE study. Ann Rheum Dis. 2012;71:198–205.PubMedCrossRef Yazici Y, Curtis JR, Ince A, Baraf H, Malamet RL, Teng LL, Kavanaugh A. Efficacy of tocilizumab in patients with moderate to severe active rheumatoid arthritis and a previous inadequate response to disease-modifying antirheumatic drugs: the ROSE study. Ann Rheum Dis. 2012;71:198–205.PubMedCrossRef
163.
Zurück zum Zitat Haraoui B, Casado G, Czirják L, Taylor A, Dong L, Button P, Luder Y, Caporali R. Tocilizumab patterns of use, effectiveness, and safety in patients with rheumatoid arthritis: final results from a set of multi-national non-interventional studies. Rheumatol Ther. 2019;6:231–43.PubMedPubMedCentralCrossRef Haraoui B, Casado G, Czirják L, Taylor A, Dong L, Button P, Luder Y, Caporali R. Tocilizumab patterns of use, effectiveness, and safety in patients with rheumatoid arthritis: final results from a set of multi-national non-interventional studies. Rheumatol Ther. 2019;6:231–43.PubMedPubMedCentralCrossRef
164.
Zurück zum Zitat Turnier JL, Brunner HI. Tocilizumab for treating juvenile idiopathic arthritis. Expert Opin Biol Ther. 2016;16:559–66.PubMedCrossRef Turnier JL, Brunner HI. Tocilizumab for treating juvenile idiopathic arthritis. Expert Opin Biol Ther. 2016;16:559–66.PubMedCrossRef
165.
Zurück zum Zitat Machado SH, Xavier RM. Safety of tocilizumab in the treatment of juvenile idiopathic arthritis. Expert Opin Drug Saf. 2017;16:493–500.PubMedCrossRef Machado SH, Xavier RM. Safety of tocilizumab in the treatment of juvenile idiopathic arthritis. Expert Opin Drug Saf. 2017;16:493–500.PubMedCrossRef
166.
Zurück zum Zitat Shepherd J, Cooper K, Harris P, Picot J, Rose M. The clinical effectiveness and cost- effectiveness of abatacept, adalimumab, etanercept and tocilizumab for treating juvenile idiopathic arthritis: a systematic review and economic evaluation. Health Technol Assess. 2016;20:1–222.PubMedPubMedCentralCrossRef Shepherd J, Cooper K, Harris P, Picot J, Rose M. The clinical effectiveness and cost- effectiveness of abatacept, adalimumab, etanercept and tocilizumab for treating juvenile idiopathic arthritis: a systematic review and economic evaluation. Health Technol Assess. 2016;20:1–222.PubMedPubMedCentralCrossRef
167.
168.
Zurück zum Zitat Nishimoto N, Kanakura Y, Aozasa K, Johkoh T, Nakamura M, Nakano S, Nakano N, Ikeda Y, Sasaki T, Nishioka K, et al. Humanized anti-interleukin-6 receptor antibody treatment of multicentric Castleman disease. Blood. 2005;106:2627–32.PubMedCrossRef Nishimoto N, Kanakura Y, Aozasa K, Johkoh T, Nakamura M, Nakano S, Nakano N, Ikeda Y, Sasaki T, Nishioka K, et al. Humanized anti-interleukin-6 receptor antibody treatment of multicentric Castleman disease. Blood. 2005;106:2627–32.PubMedCrossRef
169.
Zurück zum Zitat Kappelmann N, Lewis G, Dantzer R, Jones PB, Khandaker GM. Antidepressant activity of anti-cytokine treatment: a systematic review and meta-analysis of clinical trials of chronic inflammatory conditions. Mol Psychiatry. 2018;23:335–43.PubMedCrossRef Kappelmann N, Lewis G, Dantzer R, Jones PB, Khandaker GM. Antidepressant activity of anti-cytokine treatment: a systematic review and meta-analysis of clinical trials of chronic inflammatory conditions. Mol Psychiatry. 2018;23:335–43.PubMedCrossRef
170.
Zurück zum Zitat Taylor PC, Schiff MH, Wang Q, Jiang Y, Zhuang Y, Kurrasch R, Daga S, Rao R, Tak PP, Hsu B. Efficacy and safety of monotherapy with sirukumab compared with adalimumab monotherapy in biologic-naïve patients with active rheumatoid arthritis (SIRROUND-H): a randomised, double-blind, parallel-group, multinational, 52-week, phase 3 study. Annals of the Rheumatic Diseases. 2018;77:658–66.PubMedCrossRef Taylor PC, Schiff MH, Wang Q, Jiang Y, Zhuang Y, Kurrasch R, Daga S, Rao R, Tak PP, Hsu B. Efficacy and safety of monotherapy with sirukumab compared with adalimumab monotherapy in biologic-naïve patients with active rheumatoid arthritis (SIRROUND-H): a randomised, double-blind, parallel-group, multinational, 52-week, phase 3 study. Annals of the Rheumatic Diseases. 2018;77:658–66.PubMedCrossRef
171.
Zurück zum Zitat Zhou AJ, Lee Y, Salvadore G, Hsu B, Fonseka TM, Kennedy SH, McIntyre RS. Sirukumab: a potential treatment for mood disorders? Adv Ther. 2017;34:78–90.PubMedCrossRef Zhou AJ, Lee Y, Salvadore G, Hsu B, Fonseka TM, Kennedy SH, McIntyre RS. Sirukumab: a potential treatment for mood disorders? Adv Ther. 2017;34:78–90.PubMedCrossRef
172.
Zurück zum Zitat Sun Y, Wang D, Salvadore G, Hsu B, Curran M, Casper C, Vermeulen J, Kent JM, Singh J, Drevets WC, et al. The effects of interleukin-6 neutralizing antibodies on symptoms of depressed mood and anhedonia in patients with rheumatoid arthritis and multicentric castleman’s disease. Brain Behav. Immun. 2017;66:156–64.PubMedCrossRef Sun Y, Wang D, Salvadore G, Hsu B, Curran M, Casper C, Vermeulen J, Kent JM, Singh J, Drevets WC, et al. The effects of interleukin-6 neutralizing antibodies on symptoms of depressed mood and anhedonia in patients with rheumatoid arthritis and multicentric castleman’s disease. Brain Behav. Immun. 2017;66:156–64.PubMedCrossRef
173.
Zurück zum Zitat Wittenberg GM, Stylianou A, Zhang Y, Sun Y, Gupta A, Jagannatha PS, Wang D, Hsu B, Curran ME, Khan S, et al. Effects of immunomodulatory drugs on depressive symptoms: A mega-analysis of randomized, placebo-controlled clinical trials in inflammatory disorders. Mol Psychiatry. 2020;25:1275–85.PubMedCrossRef Wittenberg GM, Stylianou A, Zhang Y, Sun Y, Gupta A, Jagannatha PS, Wang D, Hsu B, Curran ME, Khan S, et al. Effects of immunomodulatory drugs on depressive symptoms: A mega-analysis of randomized, placebo-controlled clinical trials in inflammatory disorders. Mol Psychiatry. 2020;25:1275–85.PubMedCrossRef
174.
Zurück zum Zitat Guinane CM, Cotter PD. Role of the gut microbiota in health and chronic gastrointestinal disease: understanding a hidden metabolic organ. Therap Adv Gastroenterol. 2013;6:295–308.PubMedPubMedCentralCrossRef Guinane CM, Cotter PD. Role of the gut microbiota in health and chronic gastrointestinal disease: understanding a hidden metabolic organ. Therap Adv Gastroenterol. 2013;6:295–308.PubMedPubMedCentralCrossRef
176.
Zurück zum Zitat Cheung SG, Goldenthal AR, Uhlemann AC, Mann JJ, Miller JM, Sublette ME. Systematic review of gut microbiota and major depression. Front Psychiatry. 2019;10:34.PubMedPubMedCentralCrossRef Cheung SG, Goldenthal AR, Uhlemann AC, Mann JJ, Miller JM, Sublette ME. Systematic review of gut microbiota and major depression. Front Psychiatry. 2019;10:34.PubMedPubMedCentralCrossRef
177.
Zurück zum Zitat Kelly JR, Clarke G, Cryan JF, Dinan TG. Brain-gut-microbiota axis: challenges for translation in psychiatry. Ann Epidemiol. 2016;26:366–72.PubMedCrossRef Kelly JR, Clarke G, Cryan JF, Dinan TG. Brain-gut-microbiota axis: challenges for translation in psychiatry. Ann Epidemiol. 2016;26:366–72.PubMedCrossRef
178.
179.
Zurück zum Zitat Jiang H, Ling Z, Zhang Y, Mao H, Ma Z, Yin Y, Wang W, Tang W, Tan Z, Shi J, Li L, Ruan B. Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav Immun. 2015;48:186–94.PubMedCrossRef Jiang H, Ling Z, Zhang Y, Mao H, Ma Z, Yin Y, Wang W, Tang W, Tan Z, Shi J, Li L, Ruan B. Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav Immun. 2015;48:186–94.PubMedCrossRef
180.
Zurück zum Zitat Zheng P, Zeng B, Zhou C, Liu M, Fang Z, Xu X, Zeng L, Chen J, Fan S. Du X. Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host's metabolism. Mol Psychiatry. 2016;21:786–96.PubMedCrossRef Zheng P, Zeng B, Zhou C, Liu M, Fang Z, Xu X, Zeng L, Chen J, Fan S. Du X. Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host's metabolism. Mol Psychiatry. 2016;21:786–96.PubMedCrossRef
181.
Zurück zum Zitat Schirmer M, Smeekens SP, Vlamakis H, Jaeger M, Oosting M, Franzosa EA, Ter Horst R, Jansen T, Jacobs L, Bonder MJ, et al. Linking the human gut microbiome to inflammatory cytokine production capacity. Cell. 2016;167:1125–36 e8.PubMedPubMedCentralCrossRef Schirmer M, Smeekens SP, Vlamakis H, Jaeger M, Oosting M, Franzosa EA, Ter Horst R, Jansen T, Jacobs L, Bonder MJ, et al. Linking the human gut microbiome to inflammatory cytokine production capacity. Cell. 2016;167:1125–36 e8.PubMedPubMedCentralCrossRef
182.
Zurück zum Zitat O'Mahony L, McCarthy J, Kelly P, Hurley G, Luo F, Chen K, O'Sullivan GC, Kiely B, Collins JK, Shanahan F, et al. Lactobacillus and bifidobacterium in irritable bowel syndrome: symptom responses and relationship to cytokine profiles. Gastroenterology. 2005;128:541–51.PubMedCrossRef O'Mahony L, McCarthy J, Kelly P, Hurley G, Luo F, Chen K, O'Sullivan GC, Kiely B, Collins JK, Shanahan F, et al. Lactobacillus and bifidobacterium in irritable bowel syndrome: symptom responses and relationship to cytokine profiles. Gastroenterology. 2005;128:541–51.PubMedCrossRef
183.
Zurück zum Zitat Rinninella E, Raoul P, Cintoni M, Franceschi F, Miggiano GAD, Gasbarrini A, Mele MV. What is the healthy gut microbiota composition? A changing ecosystem across age, environment, diet. and diseases. Microorganisms. 2019;7:14.CrossRef Rinninella E, Raoul P, Cintoni M, Franceschi F, Miggiano GAD, Gasbarrini A, Mele MV. What is the healthy gut microbiota composition? A changing ecosystem across age, environment, diet. and diseases. Microorganisms. 2019;7:14.CrossRef
185.
Zurück zum Zitat Mariat D, Firmesse O, Levenez F, Guimarăes V, Sokol H, Doré J, Corthier G, Furet JP. The Firmicutes/Bacteroidetes ratio of the human microbiota changes with age. BMC Microbiol. 2009;9:123.PubMedPubMedCentralCrossRef Mariat D, Firmesse O, Levenez F, Guimarăes V, Sokol H, Doré J, Corthier G, Furet JP. The Firmicutes/Bacteroidetes ratio of the human microbiota changes with age. BMC Microbiol. 2009;9:123.PubMedPubMedCentralCrossRef
186.
Zurück zum Zitat Jeffery IB, O’Toole PW, Öhman L, Claesson MJ, Deane J, Quigley EM, Simrén M. An irritable bowel syndrome subtype defined by species-specific alterations in faecal microbiota. Gut. 2012;61:997–1006.PubMedCrossRef Jeffery IB, O’Toole PW, Öhman L, Claesson MJ, Deane J, Quigley EM, Simrén M. An irritable bowel syndrome subtype defined by species-specific alterations in faecal microbiota. Gut. 2012;61:997–1006.PubMedCrossRef
187.
Zurück zum Zitat Distrutti E, Monaldi L, Ricci P, Fiorucci S. Gut microbiota role in irritable bowel syndrome: New therapeutic strategies. World J Gastroenterol. 2016;22:2219–41.PubMedPubMedCentralCrossRef Distrutti E, Monaldi L, Ricci P, Fiorucci S. Gut microbiota role in irritable bowel syndrome: New therapeutic strategies. World J Gastroenterol. 2016;22:2219–41.PubMedPubMedCentralCrossRef
188.
Zurück zum Zitat Diviccaro S, Giatti S, Borgo F, Barcella M, Borghi E, Trejo JL, Garcia-Segura LM, Melcangi RC. Treatment of male rats with finasteride, an inhibitor of 5alpha-reductase enzyme, induces long-lasting effects on depressive-like behavior, hippocampal neurogenesis, neuroinflammation and gut microbiota composition. Psychoneuroendocrinology. 2019;99:206–15.PubMedCrossRef Diviccaro S, Giatti S, Borgo F, Barcella M, Borghi E, Trejo JL, Garcia-Segura LM, Melcangi RC. Treatment of male rats with finasteride, an inhibitor of 5alpha-reductase enzyme, induces long-lasting effects on depressive-like behavior, hippocampal neurogenesis, neuroinflammation and gut microbiota composition. Psychoneuroendocrinology. 2019;99:206–15.PubMedCrossRef
189.
Zurück zum Zitat Wang L, Christophersen CT, Sorich MJ, Gerber JP, Angley MT, Conlon MA. Increased abundance of Sutterella spp. and Ruminococcus torques in feces of children with autism spectrum disorder. Mol Autism. 2013;4:42.PubMedPubMedCentralCrossRef Wang L, Christophersen CT, Sorich MJ, Gerber JP, Angley MT, Conlon MA. Increased abundance of Sutterella spp. and Ruminococcus torques in feces of children with autism spectrum disorder. Mol Autism. 2013;4:42.PubMedPubMedCentralCrossRef
190.
Zurück zum Zitat Williams BL, Hornig M, Parekh T, Lipkin WI. Application of novel PCR-based methods for detection, quantitation, and phylogenetic characterization of Sutterella species in intestinal biopsy samples from children with autism and gastrointestinal disturbances. MBio. 2012;3:pii: e00261–11. Williams BL, Hornig M, Parekh T, Lipkin WI. Application of novel PCR-based methods for detection, quantitation, and phylogenetic characterization of Sutterella species in intestinal biopsy samples from children with autism and gastrointestinal disturbances. MBio. 2012;3:pii: e00261–11.
191.
Zurück zum Zitat Guo Q, Zheng Y, Shi J, Wang J, Li G, Li C. Immediate psychological distress in quarantined patients with COVID-19 and its association with peripheral inflammation: a mixed-method study. Brain Behav Immun. 2020;88:17–27.PubMedPubMedCentralCrossRef Guo Q, Zheng Y, Shi J, Wang J, Li G, Li C. Immediate psychological distress in quarantined patients with COVID-19 and its association with peripheral inflammation: a mixed-method study. Brain Behav Immun. 2020;88:17–27.PubMedPubMedCentralCrossRef
192.
Zurück zum Zitat Alpert O, Begun L, Garren P, Solhkhah R. Cytokine storm induced new onset depression in patients with COVID-19. A new look into the association between depression and cytokines -two case reports. Brain Behav Immun Health. 2020;9:100173.PubMed Alpert O, Begun L, Garren P, Solhkhah R. Cytokine storm induced new onset depression in patients with COVID-19. A new look into the association between depression and cytokines -two case reports. Brain Behav Immun Health. 2020;9:100173.PubMed
195.
Zurück zum Zitat Mazza MG, De Lorenzo R, Conte C, Poletti S, Vai B, Bollettini I, Melloni EMT, Furlan R, Ciceri F, Rovere-Querini P, et al. Anxiety and depression in COVID-19 survivors: Role of inflammatory and clinical predictors. Brain Behav Immun. 2020;89:594–600.PubMedPubMedCentralCrossRef Mazza MG, De Lorenzo R, Conte C, Poletti S, Vai B, Bollettini I, Melloni EMT, Furlan R, Ciceri F, Rovere-Querini P, et al. Anxiety and depression in COVID-19 survivors: Role of inflammatory and clinical predictors. Brain Behav Immun. 2020;89:594–600.PubMedPubMedCentralCrossRef
196.
Zurück zum Zitat Ahmed MZ, Ahmed O, Aibao Z, Hanbin S, Siyu L, Ahmad A. Epidemic of COVID-19 in China and associated psychological problems. Asian J Psychiatr. 2020;51:10209.CrossRef Ahmed MZ, Ahmed O, Aibao Z, Hanbin S, Siyu L, Ahmad A. Epidemic of COVID-19 in China and associated psychological problems. Asian J Psychiatr. 2020;51:10209.CrossRef
198.
Zurück zum Zitat Coomes EA, Haghbayan H. Interleukin-6 in Covid-19: a systematic review and meta- analysis. Rev Med Virol. 2020;30:1–9.PubMedCrossRef Coomes EA, Haghbayan H. Interleukin-6 in Covid-19: a systematic review and meta- analysis. Rev Med Virol. 2020;30:1–9.PubMedCrossRef
199.
Zurück zum Zitat Mojtabavi H, Saghazadeh A, Rezaei N. Interleukin-6 and severe COVID-19: a systematic review and meta-analysis. Eur Cytokine Netw. 2020;31:44–9.PubMedPubMedCentralCrossRef Mojtabavi H, Saghazadeh A, Rezaei N. Interleukin-6 and severe COVID-19: a systematic review and meta-analysis. Eur Cytokine Netw. 2020;31:44–9.PubMedPubMedCentralCrossRef
200.
Zurück zum Zitat Sun H, Guo P, Zhang L, Wang F. Serum Interleukin-6 Concentrations and the Severity of COVID-19 Pneumonia: A Retrospective Study at a Single Center in Bengbu City, Anhui Province, China. in January and February 2020. Med Sci Monit. 2020;26:e926941.PubMed Sun H, Guo P, Zhang L, Wang F. Serum Interleukin-6 Concentrations and the Severity of COVID-19 Pneumonia: A Retrospective Study at a Single Center in Bengbu City, Anhui Province, China. in January and February 2020. Med Sci Monit. 2020;26:e926941.PubMed
201.
Zurück zum Zitat Gubernatorova EO, Gorshkova EA, Polinova AI, Drutskaya MS. IL-6: relevance for immunopathology of SARS-CoV-2. Cytokine Growth Factor Rev. 2020;53:13–24.PubMedPubMedCentralCrossRef Gubernatorova EO, Gorshkova EA, Polinova AI, Drutskaya MS. IL-6: relevance for immunopathology of SARS-CoV-2. Cytokine Growth Factor Rev. 2020;53:13–24.PubMedPubMedCentralCrossRef
202.
Zurück zum Zitat Liu B, Li M, Zhou Z, Guan X, Xiang Y. Can we use interleukin-6 (IL-6) blockade for coronavirus disease 2019 (COVID-19)-induced cytokine release syndrome (CRS)? J Autoimmun. 2020:102452. Liu B, Li M, Zhou Z, Guan X, Xiang Y. Can we use interleukin-6 (IL-6) blockade for coronavirus disease 2019 (COVID-19)-induced cytokine release syndrome (CRS)? J Autoimmun. 2020:102452.
203.
204.
Zurück zum Zitat Benedetti F, Mazza M, Cavalli G, Ciceri F, Dagna L, Rovere-Querini P. Can cytokine blocking prevent depression in COVID-19 survivors? J Neuroimmune Pharmacol. 2020:1–3. Benedetti F, Mazza M, Cavalli G, Ciceri F, Dagna L, Rovere-Querini P. Can cytokine blocking prevent depression in COVID-19 survivors? J Neuroimmune Pharmacol. 2020:1–3.
205.
Zurück zum Zitat Catanzaro M, Fagiani F, Racchi M, Corsini E, Govoni S, Lanni C. Immune response in COVID-19: addressing a pharmacological challenge by targeting pathways triggered by SARS-CoV-2. Sig Transduct Target Ther. 2020;5:84.CrossRef Catanzaro M, Fagiani F, Racchi M, Corsini E, Govoni S, Lanni C. Immune response in COVID-19: addressing a pharmacological challenge by targeting pathways triggered by SARS-CoV-2. Sig Transduct Target Ther. 2020;5:84.CrossRef
Metadaten
Titel
On inflammatory hypothesis of depression: what is the role of IL-6 in the middle of the chaos?
verfasst von
Elnaz Roohi
Nematollah Jaafari
Farshad Hashemian
Publikationsdatum
01.12.2021
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2021
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-021-02100-7

Weitere Artikel der Ausgabe 1/2021

Journal of Neuroinflammation 1/2021 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie

„Übersichtlicher Wegweiser“: Lauterbachs umstrittener Klinik-Atlas ist online

17.05.2024 Klinik aktuell Nachrichten

Sie sei „ethisch geboten“, meint Gesundheitsminister Karl Lauterbach: mehr Transparenz über die Qualität von Klinikbehandlungen. Um sie abzubilden, lässt er gegen den Widerstand vieler Länder einen virtuellen Klinik-Atlas freischalten.

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Was nützt die Kraniektomie bei schwerer tiefer Hirnblutung?

17.05.2024 Hirnblutung Nachrichten

Eine Studie zum Nutzen der druckentlastenden Kraniektomie nach schwerer tiefer supratentorieller Hirnblutung deutet einen Nutzen der Operation an. Für überlebende Patienten ist das dennoch nur eine bedingt gute Nachricht.

ADHS-Medikation erhöht das kardiovaskuläre Risiko

16.05.2024 Herzinsuffizienz Nachrichten

Erwachsene, die Medikamente gegen das Aufmerksamkeitsdefizit-Hyperaktivitätssyndrom einnehmen, laufen offenbar erhöhte Gefahr, an Herzschwäche zu erkranken oder einen Schlaganfall zu erleiden. Es scheint eine Dosis-Wirkungs-Beziehung zu bestehen.