Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 7083 2023-01-31 00:07:22 |
2 format -1 word(s) 7082 2023-01-31 03:52:56 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Harsanyi, S.;  Kupcová, I.;  Danisovic, L.;  Klein, M. Cytokines as Selected Biomarkers of Depression. Encyclopedia. Available online: https://encyclopedia.pub/entry/40626 (accessed on 20 April 2024).
Harsanyi S,  Kupcová I,  Danisovic L,  Klein M. Cytokines as Selected Biomarkers of Depression. Encyclopedia. Available at: https://encyclopedia.pub/entry/40626. Accessed April 20, 2024.
Harsanyi, Stefan, Ida Kupcová, Lubos Danisovic, Martin Klein. "Cytokines as Selected Biomarkers of Depression" Encyclopedia, https://encyclopedia.pub/entry/40626 (accessed April 20, 2024).
Harsanyi, S.,  Kupcová, I.,  Danisovic, L., & Klein, M. (2023, January 31). Cytokines as Selected Biomarkers of Depression. In Encyclopedia. https://encyclopedia.pub/entry/40626
Harsanyi, Stefan, et al. "Cytokines as Selected Biomarkers of Depression." Encyclopedia. Web. 31 January, 2023.
Cytokines as Selected Biomarkers of Depression
Edit

Depression is one of the leading mental illnesses worldwide and lowers the quality of life of many. According to WHO, about 5% of the worldwide population suffers from depression. Studies report a staggering global prevalence of 27.6%, and it is rising. Professionally, depression belonging to affective disorders is a psychiatric illness, and the category of major depressive disorder (MDD) comprises various diagnoses related to persistent and disruptive mood disorders. Due to this fact, it is imperative to find a way to assess depression quantitatively using a specific biomarker or a panel of biomarkers that would be able to reflect the patients’ state and the effects of therapy. Cytokines, hormones, oxidative stress markers, and neuropeptides are studied in association with depression. 

depression cytokines interleukins chemokines

1. Interleukin-1 Beta

Interleukin-1 beta (IL-1β) is a pro-inflammatory cytokine encoded by the IL1B gene, located at 2q14.1. This cytokine was first implicated in the etiology of sickness behavior and later linked to depressive disorder along with IL-6 and TNF [1][2]. Elevated levels of various inflammatory markers have been found in episodes of depressive behavior. However, the elevation of pro-inflammatory cytokines is primarily a reaction to acute or chronic stress, where IL-1β was found to be elevated in 38.5% and 75.6% of studies, respectively [3][4]. Moreover, danger signals are one of the inducers of the inflammatory response. This happens via inflammasomes or toll-like receptors (TLRs) and leads to elevated IL-1β, IL-6, and TNF-α [5][6]. Untreated patients suffering from depression have been found to have elevated IL-1β levels in CSF. This raises the question of whether this occurrence is more related to depression or acute reaction to stress, as mice under stress expressed more IL-1β than controls [7][8]. This association was later studied by Thomas et al., who found that elderly subjects (above 60 years) expressed IL-1β proportionally to illness severity [9]. Corwin et al. reported elevated IL-1β levels in women suffering from postpartum depression (PPD) [10]. A meta-analysis concluded that IL-1β was significantly elevated in patients with depression and Alzheimer’s disease (AD), while IL-6 was elevated only in patients with depression [3]. Recently, Ferreira et al. reported a possible association of SNP in the IL1B gene (rs16944; 511 C > T) with higher susceptibility to depression in patients with multiple sclerosis (MS) [11].
Ovaskainen et al. studied IL-1β in males and concluded that decreased levels of this cytokine and elevated levels of the interleukin-1 receptor antagonist (IL-1RA) were more frequent in depressed subjects [12]. These findings contradict each other even if there is a sex difference. Noteworthy, however, is the relation between IL-1β and the P2X family of receptors, in which P2X7 inhibition has been associated with antidepressant-like effects, and IL-1β knockout mice failed to express these receptors [13][14]. Stress-induced production of pro-inflammatory cytokines was reported by Portfield et al., where repeated stressor exposure in rats enhanced the production of IL-1β via the norepinephrine beta-adrenergic pathway [15]. However, a subsequent study by Barnard et al. reported only male rats to be affected by the beta-adrenergic pathway, while females remained unaffected [16]. The use of intrahippocampal IL-1RA causes downregulation of brain-derived neurotrophic factor (BDNF) in individuals experiencing social isolation [17]. IL-1RA modulated synaptic plasticity in a rat AD model, where acute beta-amyloid mediated potentiation impairment was possibly caused by inflammatory response and release of IL-1β [18]. IL-1Ra knockout mice show anxiety-like behavior as they grow older [19]. Receptor upregulation has been found in patients suffering from migraine with aura symptoms, believed to be associated with cortical spreading depression [20]. This upregulation may be a response to modulate the inflammatory process.

2. Soluble Interleukin-2 Receptor

The soluble IL-2 receptor (sIL-2R) is approx. 40 kDa in size and results from proteolytic cleavage of the ectodomains of the membrane-bound IL-2Rα chain as a result of T cell activation. Its levels are increased in inflammation, infections, and autoimmune disease. sIL-2R has a low affinity for IL-2 and can function either as a decoy receptor reducing the bio-availability of IL-2 or by enabling trans presentation to cells expressing the dimeric, intermediate-affinity IL-2R, which leads to enhanced or suppressed immune response depending on the type of cells involved [21][22]. The role sIL-2R may play in depression was first mentioned in the literature in 1991 in an article exploring depression-related disturbances in mitogen-induced lymphocyte responses together with the production of IL-1β and sIL-2R, suggesting that T cell activation may be one of the characteristics of MDD [23]. Serum levels of sIL-2R are increased in MDD and correlate with somatoform symptoms and somatic anxiety in depression but not with the overall severity of the disease [21][24]. That serum sIL-2R was increased in MDD was later reaffirmed, with the mean difference in sIL-2R levels between healthy subjects and MDD patients being 0.555 U/mL [25].

3. Interleukin-3

Interleukin-3 (IL-3) is a growth-promoting cytokine encoded by the IL3 gene located at 5q31.1 [26]. It is a multipotent hematopoietic growth factor expressed mainly by activated T-lymphocytes and macrophages inducing proliferation, maturation, and perhaps self-renewal of stem cells committed to the hematopoietic lineage [27]. The relationship between serum IL-3 and psychiatric conditions was reported by Xiu et al., who focused on schizophrenic patients. Compared to healthy controls, the study group had significantly elevated IL-3 levels. Moreover, IL-3 levels were positively correlated with depressive sub-scores [28]. The most recent 2022 paper by Gao et al. compared serum cytokine levels, including IL-3, in normal and overweight patients with their first depressive episode without the medication history of taking antidepressants. Of the 37 cytokines measured, IL-3 was significantly elevated in overweight, depressed patients compared to their normal-weight depressed counterparts [29]. These results indicate that these two inflammation-related conditions might share some aspects of their pathogenesis. A 2020 meta-analysis by Osimo et al. analyzed 107 studies comprising 5166 patients and 5083 controls and found IL-3 among those cytokines whose blood levels were significantly elevated in depressed subjects [30]. Overall, IL-3 seems to be another valuable marker of depression, despite there not being much information about its precise action.

4. Interleukin-4

Interleukin-4 (IL-4) is coded by the IL4 gene, a 160 kb region on the long arm of chromosome 5. IL-4 plays a role in the regulation of IgE synthesis, differentiation of naive T cells into the Th2 subtype, suppresses IL-2 production, and promotes the expression of anti-inflammatory cytokines such as TGF-β1. IL-4 is produced by CD4 T cells, basophils, eosinophils, mastocytes, NK cells, and innate lymphoid cells type 2, while production in CNS by microglia and neuronal cells [31][32][33]. Its effect on the brain is of particular importance for microglia specialization. Microglia exposed to IL-4 and IL-13 specialize to the M2 anti-inflammatory subtype in contrast to the pro-inflammatory M1 subtype induced by exposure to IFN-α, TNF-β, or lipopolysaccharide (LPS). IL-4-driven Arg1+ microglia in the hippocampus trigger brain-derived neurotrophic factor (BDNF)-driven neurogenesis in reaction to stress, resulting in improved resilience to stress-induced depression. In a murine model, mice were exposed to chronic mild stress, and subsequently, levels of IL-4 and other cytokines in their hippocampi were measured using an immunofluorescence assay. The results showed overexpression of IL-4 in the mice group with low-stress susceptibility [34][35]. IL-4 inhibits the serotonin transporter activity in a dose-dependent manner, hence increasing the amount of serotonin available, acting similarly to some of the currently used antidepressants. This is supported by the fact that the use of an anti-IL-4 antibody reversed the effect IL-4 had on serotonin transporter levels in an experiment using B lymphoblasts. In a later animal model, rodents treated with IFN-α showed more depressive behaviors in the forced swim test, tail suspension test, and active avoidance task when their IL-4 microglial reactivity was reduced [35][36]. In agreement with these findings, IL-4−/− mice tested in the forced swim test and tail suspension test showed significant depression-like behaviors compared to wild-type rodents, which was not further increased by the application of IFN-α [37].

5. Interleukin-5

Interleukin-5 (IL-5) is coded by a gene clustered with IL-3 and IL-4 on chromosome 5. It is involved in the activation, proliferation, and differentiation of eosinophils and is best known for its involvement in the pathophysiology of autoimmune disorders such as asthma and Graves’ disease [38][39]. IL-5 is produced by activated Th2 cells, microglia, mastocytes, and astrocytes; has mitogenic effects on microglia; and activates JAK2 and STAT5 pathways involved in cell proliferation, which can lead to depressive symptoms via glucocorticoid signaling. IL-5 is also involved in Ras-ERK pathways, the hyperactivity of which causes deficits in synaptic plasticity and hippocampal-related learning in mice [39][40][41]. Although the role of IL-5 in autoimmune and parasitic diseases is well described, little is known about its potential involvement in depressive disorder, and the findings have been inconsistent so far. Elomaa et al. described elevated IL-5 levels in 58 patients with MDD, though a later meta-analysis did find changes in levels of IL-5 to be inconclusive in depressed patients. Recent research on circulating cytokine levels in breast cancer patients even found IL-5 levels decreased in the depressed group [42][43].

6. Interleukin-6

The gene encoding interleukin-6 (IL-6) is located on chromosome 7, containing five exons [44]. IL-6 is released by Th1 and Th2 cells, having either pro- or anti-inflammatory properties, depending on the presence of either the IL-6 receptor or the membrane-bound gp130 signal transducer, which are expressed within specific cell types throughout the body. Microglia secrete IL-6 by interactions of SDF-1 with CXCR4, leading to enhanced IL-6 mRNA expression and increased protein synthesis by activating the MAPK/ERK signal pathway and phosphatidylinositol-3 kinase. Within the peripheral and central nervous systems, IL-6 can act as a neuronal growth factor leading to neurite development and nerve regeneration. In reaction to stressful stimuli, the synthesis of IL-6 increases, and findings suggest that in depression, higher IL-6 levels correlate with a more severe course of the disease [45][46][47]. It also seems to be linked to specific symptoms or subtypes of depressive disorder, as the relationship between IL-6 levels and reduced appetite, sleep disturbances, low mood, and feelings of worthlessness has been confirmed [48]. Recently, Li et al., studied peripheral IL-6 levels and changes in white matter for two years after stressful life events (SLEs) in 185 subjects using diffusion tensor imaging to elucidate its role in developing depression after SLEs. The participants exhibited high levels of IL-6 during the stress period and a corresponding decline in IL-6 levels as the stress period ended. IL-6 correlated with the overall level of stress. After the period of acute stress, the IL-6 white matter network differences were shown to be strongly associated with the interindividual variation in susceptibility to depressive disorder in healthy individuals with SLEs [46]. However, it is not yet clear if the elevated plasma levels of IL-6 also translate to the same changes within the brain and if the elevated IL-6 level is a cause or consequence of depressive symptoms, as the results regarding the correlation between plasma and CSF IL-6 levels vary. In animal models of depression, IL-6 was reduced in the CA1 region of the hippocampus of rats exhibiting depressive behaviors and IL-6 knockdown facilitated development of depressive behaviors. At the same time, overexpression of IL-6 led to a reduction in these behaviors [49][50]. These conflicting findings could mean that there is a more complex relationship between plasmatic and CSF levels of IL-6, that IL-6 is released after stressors in a neuroprotective fashion, or they could point to a different role of IL-6 in rodent models of depression compared to human patients with depressive disorder.
Soluble IL-6 receptor (sIL-6R) results from alternative mRNA splicing, proteolysis of the membrane-bound IL-6R, and the release of extracellular vesicles as it is involved in the IL-6 pro-inflammatory trans-signaling. It is the only IL-6 receptor type in astrocytes and neurons [51][52]. In addition to increased IL-6 levels, sIL-R6 levels were reported in depressed patients, with elevated levels of both persisting even after the start of antidepressant treatment [53][54]. Contrary to these findings, sIL-6R was decreased in the CSF of elderly depressed patients medicated with antidepressants in combination with neuroleptics or benzodiazepines [55]. The reasons for these inconsistencies are yet to be further explored.

7. Interleukin-8

Interleukin-8 (IL-8 or CXCL8) is a chemokine produced by, e.g., macrophages, microglia, or endothelial cells. The CXCL8 gene codes IL-8 with a cytogenetic location of 4q13.3. Microglial IL-8 reacts to pro-inflammatory stimuli, and elevation in the periphery has been linked to schizophrenia, bipolar disorder, or autism spectrum disorders. At the same time, its connection to MDD is relatively new and inconsistent [56]. Inconsistencies start with sex differences in rats, where the IL-8 effect on disease severity was associated only with females [57]. Female rats with lower IL-8 levels are more susceptible to medication and electroconvulsive therapy (ECT) [58][59]. In human studies, breast cancer survivors (100% female) with higher basal IL-8 levels at the start of the study were less likely to experience recurrent depression [60]. However, inconsistencies continue, as in a study on geriatric depression (patients 70–84 years of age), higher levels of IL-8 and IL-6 in CSF were associated with current depression [61]. Elevated levels of IL-8 in CSF were also observed by Kuzior et al. in patients suffering from unipolar depression [62]. Whole blood sample examination in a randomized study reported the possibility of IL-8 effect on depression to be rather mitigative than enhancing, where patients with higher levels of IL-8 are less likely to suffer from inflammation-associated depression [63]. Partly in line with previous studies is a study on IL-8 in delirium, dementia, and depression by Sajjad et al., who found an increased IL-8 concentration in patients with delirium/depression. In contrast, the group with delirium/dementia expressed lower levels of IL-8 [64].

8. Interleukin-9

The gene for interleukin-9 (IL-9) is located on chromosome 5 within the TH2 cytokine cluster in the region q31–35, consisting of five exons and four introns. Though initially believed to be a Th2 cytokine, it was recently revealed that it is copiously secreted by newly discovered Th9 cells. The binding of IL-9 with its receptor promotes cross phosphorylation of Janus kinase (JAK) 1 and JAK3, leading to the activation of signal transducer and activator of transcription (STAT) 1, 3, and 5 [65][66]. IL-9 has also been implicated in the pathophysiology of depression. Upregulation of the IL9 gene was found in the post-mortem analysis of the brains of patients with MDD in the Brodman Area 10—a part or brain involved in the mediation of reward-related behavior. Elevated IL-9 levels were found to correlate with maternal mid-pregnancy symptoms of depression and anxiety [67][68]. Varshney et al., found elevated blood IL-9 levels in depressed patients with type 2 diabetes compared to diabetic patients without depressive symptoms [69]. Elevated IL-9 was also reported in the saliva of MDD patients. However, it is not yet thoroughly researched to what degree the changes in saliva correlate with changes in blood samples [70].

9. Interleukin-10

Interleukin-10 is coded by the IL10 gene located on chromosome 1 (1q31–32) and contains five exons. By acting on dendritic cells (DCs) and macrophages, IL-10 inhibits the development of TH1- and TH2-type responses, acting as an anti-inflammatory cytokine [71][72]. IL-10 knockout mice showed learned helplessness in animal depression models, a symptom that could be reverted by application of IL-10, whereas IL-10 administration led to increased motor activity in wild-type mice. Decreased levels of IL-10 were described in humans with symptoms of depression, anxiety, and increased suicide risk in a population study, as well as in stroke patients who developed the depressive disorder. Yet the literature also yields studies that report increased IL-10 levels in MDD. One suggested explanation for this discrepancy is that IL-10 levels increase initially in response to acute inflammation connected with depression as a part of the compensatory immune system. However, if the inflammation is not successfully attenuated with a longer disease duration, the IL-10 levels eventually decrease [73][74][75][76]. This theory is also supported by the fact that several types of antidepressants increase IL-10 levels [77][78].

10. Interleukin-12

Interleukin-12 (IL-12) is a heterodimeric pro-inflammatory cytokine encoded by two genes for each subunit, IL12A and IL12B, located at 3q25.33 and 5q33.3, respectively [79][80]. It was first discovered and described in the 1980s as a result of experiments with cell culture of Epstein-Barr virus-transformed lymphoblastoid cell lines. The heterodimeric biologically active form is also referred to as IL-12p70 [81]. It is synthesized mainly by antigen-presenting cells (APCs) such as macrophages or DCs. Its primary function is to help combat intracellular infectious agents, and it can be mobilized within minutes [82]. It is a highly potent inducer of Th1 response in humans [83]. The potential role of IL-12/IL-12p70 (from now on, it will be referred to as just IL-12) in autoimmunity and its relation to the etiopathogenesis of neurodevelopmental disorders such as autism was first studied 26 years ago. Singh performed a study on randomly selected 20 autistic children. The study group had significantly higher plasma levels of IL-12 and IFN-γ [84]. They scrutinized the correlation and possible causation between plasma level of IL-12 and neuropsychiatric disorders, including MDD, bipolar disorder, and schizophrenia. Kim et al., published a comparative study recruiting 102 psychiatric patients, 34 of whom had MDD. The two principal findings were that, first, the plasma levels of IL-12 were significantly higher in patients with depression compared to controls, and second, there was a significant decrease in IL-12 plasma levels after eight weeks of antidepressant pharmacotherapy [85]. Lee and Kim reproduced similar results in a clinical trial [86]. Along the same lines, Sutcigil et al. found elevated serum levels of IL-12, which decreased after eight weeks of sertraline therapy [87]. A recent 2019 study presented a similar picture. Nowak et al. found that patients with MDD without any comorbidities had higher plasma levels of IL-12, which can be mitigated by a sub-anesthetic dose of ketamine which acts through the promotion of M2 phenotype in macrophages [88]. El-Tantawy et al. chose an original approach that investigated serum levels of a cytokine palette, including IL-12, in patients diagnosed with rheumatoid arthritis (RA). The main aim was to correlate the cytokine levels with psychiatric disorders, mainly depressive disorder, and anxiety, which are more common in RA patients. Again, the levels of IL-12 were substantially elevated in the study group [89]. Schmidt et al. compared the serum levels of nine different cytokines, including IL-12. They originally designed the study so that the cytokine levels in depressive disorder were put into perspective with other immune system-associated conditions, such as obesity. Unsurprisingly, IL-12 was elevated in depressed patients. Comparing the obese and non-obese subgroups, both had IL-12 significantly elevated. However, from the whole spectrum of studied pro-inflammatory cytokines, more were elevated in the non-obese group. However surprising it might seem at first glance, it can point to the fact that obesity causes the elevation of pro-inflammatory status by itself, independent of depression. Thus, in the case of depression, only a few cytokines are elevated compared to the baseline chronic pro-inflammatory status. Therefore, obesity can mask depression-induced cytokine alteration, making the cytokine level results harder to interpret. Strikingly, the authors also found that IL-12 was elevated in the unemployed and retired [90]. The effect of serotonin re-uptake inhibitor (SSRI) treatment in patients with generalized anxiety disorder and its influence on IL-12 and other cytokine levels was the principal focus of the 2019 study by Hou et al., who reported a significant reduction in anxiety and pro-inflammatory cytokines levels compared to baseline figures [91]. All these results have been corroborated by a 2017 meta-analysis which included 82 studies, underlining the reproducibility of IL-12 plasma/serum level elevation in depressed patients [42]. On the other hand, a 2020 systematic review and meta-analysis investigated whether the examination of baseline levels of peripheral cytokines can predict the responsiveness to the antidepressant treatment. There was no difference in IL-12 (plus ten other cytokines) levels between subsequent responders and non-responders [92]. Taken together, the investigation of IL-12 serum/plasma level can be clinically valuable within a complex diagnostic approach. However, it always has to be interpreted with caution since many other conditions, e.g., obesity, can alter the same pro-inflammatory pathways, overlapping with each other.

11. Interleukin-13

Interleukin-13 (IL-13), first discovered in 1993, is a cytokine encoded by the IL13 gene at located 5q31.1 [93]. It is closely associated with the IL4 gene (5q23-31), and the two gene products (IL-13 and IL-4) also share some functional characteristics [94]. IL-13 is generally defined as an anti-inflammatory cytokine. For instance, it can mitigate the pro-inflammatory activation of microglia in the brain, providing neuroprotection during ischemic stroke. However, its effects can be pleiotropic, so under certain circumstances, IL-13 can contribute to or fail to suppress pro-inflammatory responses [95][96][97]. Corren reviewed that IL-13 plays a central role in various pathogenetic aspects of allergic conditions, such as allergic asthma, including the B lymphocyte switch to IgG production [98]. Thus, it is primarily associated with Th2-mediated immune functions [99]. Discussing the role of IL-13 in psychiatric conditions, Pavón et al. found elevated IL-13 serum levels in MDD patients. The most probable explanation of this finding was that chronic MDD-associated stress causes the chronic elevation of cortisol, which can raise IL-13 in peripheral blood [100]. The relationship between neuroendocrine-immune regulation and stress is complex, including Th1 and Th2 responses, depending on the stress duration. While acute stress is Th1-mediated and can be beneficial, chronic stress elicits deleterious Th2 responses, which may disrupt immune system integrity [101]. Hernandéz et al. authored a controlled clinical trial focused on the effect of SSRI treatment on various cytokine levels in MDD patients. At week 0, the depressed subjects had significantly elevated serum levels of IL-13, with a staggering 69% reduction at week 52 [102]. Wong et al., reported conflicting results of decreased levels of IL-13 in MDD patients. They discussed that such discrepancy might have arisen due to differences in gender, age, or severity of the disorder [103]. Czysz et al., compared 19 inflammatory markers as potential MDD treatment prediction modeling tools. Only IL-13 was found to be able to predict differential treatment outcomes [104]. The above-cited meta-analysis by Köhler et al. underpins the association between peripheral IL-13 elevation and MDD [105]. Despite acting on the opposite side of the Th1/Th2 dichotomy, IL-13 is associated with MDD similarly to IL-12. In a recent 2021 study, Vai et al. attempted to investigate the relationship between inflammatory profile and suicide and found that plasmatic IL-13 was elevated in patients with a history of suicidal behavior [106]. The link between IL-13 and suicidal behavior was also presented by Tonelli et al., who examined the samples of orbitofrontal cortices, namely the Brodmann area 11 of suicide victims. One of the selection criteria for the particular cytokine panel investigated, which included IL-13, was the previously published association between allergies and depression or even suicide [107][108]. The investigation revealed an increased expression of IL-13 in this area, typically associated with decision-making and planning [109].

12. Interleukin-17A

Interleukin-17A (IL-17A) is a pro-inflammatory cytokine encoded by the IL17A gene located at 6p12.2 [110]. A member of the highly evolutionarily conserved IL-17 cytokine superfamily, it was first discovered in 1993 [111]. Later, IL-17A was revealed to be the signature cytokine of a specific population of CD4+ T cells, known as T helper 17 or Th17 cells. Over the almost 30 years after its initial cloning, many research teams found it to be more than just a pro-inflammatory cytokine. McGeachy et al. reviewed its physiological functions, which include wound healing, extracellular matrix remodeling, role in thermogenesis, and others. From the pathological perspective, IL-17A has been implicated in the pathogenesis of autoimmune diseases, obesity-associated chronic low-grade inflammation, tumorigenesis, neurodegeneration, depression, and autism [112]. Matusevicius et al. found that blood and CSF from patients with MS contained, compared to controls, higher numbers of IL-17A-expressing mononuclear leucocytes [113]. In 2021, Nothdurfer et al. observed that MDD non-responders to antidepressant therapy had significantly elevated IL-17A levels, supporting its clinical relevance as a marker of therapy resistance [114]. Medina-Rodriguez et al. analyzed stool samples taken from mice to investigate the potential relation between dysbiosis of the gut microbiome and MDD. They found a relationship between certain types of gut microorganisms and the development of depression-like symptoms. The primary cells which mediated these processes were IL-17A-producing Th17 cells [115]. Alvarez-Mon et al. enrolled 30 MDD patients and 30 control subjects and compared the Th1, Th2, and Th17 subsets of CD4+ T cells. The study data showed that the circulating Th17 cell count was higher with corresponding increased serum levels of IL-17-A. The IL-17A overexpression was observed in various activation/differentiation stages of CD4+ T cells, namely the naïve, central memory, and effector memory T cells [116]. Min et al. studied the role of IL-17A in developing PPD and found a link between the elevated peripheral levels of IL-17A and the risk of PPD development [117]. The bottom line is that IL-17A positively correlates with MDD and other related conditions and can be used in diagnostic practice and therapy success prediction.

13. Interleukin-18

Interleukin-18 is a pro-inflammatory cytokine encoded by the IL18 gene located at 11q23.1. Belonging to the IL-1 superfamily, IL-18 can stimulate both the Th1 and Th2 lines of differentiation [118]. Its discovery dates back to 1989, when Nakamura et al., described an “unidentified” factor capable of stimulating IFN-γ production, only later on designated as IL-18, or based on its first known action, as an interferon-gamma inducing factor [119]. In 2005, IL-18 was already established as a key player in neuroinflammation and neurodegeneration, as reviewed by Felderhoff-Mueser et al. [120]. IL-18 (along with IL-1β) is associated with inflammasomes—cytosolic multiprotein complexes, which, upon stimulation, induce the synthesis of these two pro-inflammatory cytokines by the enzymatic action of caspases. Inflammasome activation can also lead to different types of cell death, including apoptosis and pyroptosis [121]. Studying the inflammasome pathways and their supposed relation to MDD, Wong et al. focused on genetically and pharmacologically induced inhibition of caspase-1 and found that both genetic suppression and minocycline-induced antagonism of this inflammasome-associated enzyme ameliorated depressive and anxiety-like behavior in mice. They also found that minocycline had a beneficial effect on the gut microbiome, hypothesizing about the presumed interconnection known as the microbiota–inflammasome–brain axis [122]. In 2020, Song et al. established a mice model of chronic stress to investigate the potential role of NLRP1 inflammasome in the pathogenesis of MDD. The results showed that chronic stress activated NLRP1 inflammasome in the hippocampus, which in turn induced the release of pro-inflammatory cytokines, including IL-18. The mice with knocked down NLRP1 inflammasome activity displayed diminished chronic stress-induced depressive-like demeanor, indicating that inflammasome-mediated IL-18-induced CNS inflammation can indeed lead to depression [123]. Interestingly, Fan et al., examined the serum levels of three cytokines, one of which was IL-18, but found IL-18 to be lower in depressed subjects. They hypothesized that the results might reflect that the patients were treated with antidepressants, which could have lowered the serum levels of IL-18 [124]. Controlling for this variable, Kokai et al. measured the serum levels of IL-18 in 13 patients. Eight were diagnosed with MDD and five with panic disorder, but none were taking any psychotropic medications. In this scenario, IL-18 serum levels were significantly elevated [125]. Alcocer-Goméz et al. directly compared the differences in IL-18 levels between 20 “no treatment” subjects and 20 treated with amitriptyline, examining NLRP3 inflammasome and caspase-1 activity. Following the previously cited studies, the expression of NLRP3 and caspase-1-associated genes was increased, correlating with the elevation of IL-18 serum levels [126]. Prossin et al. implemented an innovative approach and correlated the IL-18 plasma levels with positron emission tomography (PET)-visualized µ-opioid receptor activity using the selective radiotracer [11C] carfentanil. Unsurprisingly, baseline IL-18 values were higher in MDD subjects, but the most exciting finding was that the IL-18 rose after sadness induction in healthy subjects, correlating with µ-opioid receptor activity [127]. All in all, IL-18 serum/plasma levels seem to correlate with IL-18 action as a potent activator of MDD-associated inflammatory changes in the CNS, which can be used diagnostically.

14. Interferon-Gamma

Interferon-gamma (IFN-γ) is encoded by the IFNG gene located at 12q15. IFN-γ plays a role in stress-induced immune dysregulation [128]. Women with higher subjective stress after breast cancer surgery but before adjuvant therapy were found to have lower basal and IFN-γ-augmented NK cells and reduced T cell proliferative response to mitogens [129][130]. Additionally, antidepressants were found to suppress the release of IFN-γ by activated T cells [131]. Elevated production of IFN-γ by CD3+ CD4+ T lymphocytes was observed in fatigued and depressed patients suffering from MS compared to non-fatigued MS patients [132]. The effect of IFN-γ on mood abnormalities via the cannabinoid CB1 receptor pathway has been observed in mice [133]. A meta-analysis on cytokines in MDD reported measurements extracted from research articles including 131 depressed and 107 nondepressed subjects, in which concentrations of IFN-γ did not differ between these groups [134]. However, a different study observed significantly decreased serum IFN-γ levels in patients with MDD compared to healthy controls [135]. While decreased in base levels, the reactivity to mitogens in depressive patients is significantly enhanced [136][137]. A different take on IFN-γ by Zhang et al. studied microglia isolated from the hippocampus of IFN-γ-injected mice, where the proliferation of neural stem/precursor cells (NSPCs) was suppressed by the inhibition of the JAK/STAT1 pathway [138]. The IFN-γ injection led to impairment of adult hippocampal neurogenesis and led to cognitive defects and depression-like behavior [138]. IFN-γ signaling was reported to play a role in myelin disruption in patients with migraine with aura and depression [139]. Moreover, a genetic variant (+874) T/A) has been proposed as a possible risk factor for patients, as they could be predisposed to IFN-induced depression [140]. A recent review of in vitro and ex vivo studies on the effects of different classes of antidepressant drugs reported that many of these drugs prevent IFN-γ microglial activation [141]. The effect of IFN-γ polymorphism on tryptophan metabolism and therapy was studied by Myint et al., who reported that the presence of the IFN-γ CA-repeat allele 2 (homozygous) resulted in significantly lower serum tryptophan and 5-hydroxy indole acetic acid (5HIAA). In contrast, serum kynurenine was considerably higher [142]. These findings point to a strong link between IFN-γ and depression.

15. CCL2

The chemokine (C-C motif) ligand 2 (CCL2) is a member of a chemokine superfamily characterized by the N-terminal cysteine arrangement variability comprising four subfamilies. The CC chemokine subfamily is represented by those chemokines which contain two adjacent cysteine residues, hence the name. CCL2 is encoded by the CCL2 gene located at 17q12, along with several other cytokines clustered in this location [143]. It is a pro-inflammatory cytokine secreted mainly by APCs, e.g., DCs cells with chemoattractant properties towards several populations of white blood cells, including the representatives of innate and adaptive immune systems alike. The principal cell targets attracted by chemotaxis towards the CCL2 signals are monocytes, giving it the alternative designation—monocyte chemoattractant protein-1 (MCP-1) [144]. Recently, Proma et al. found that CCL2/MCP-1 was decreased in the serum of MDD patients [145]. Myung et al. reported a similar association between MDD and low CCL2/MCP-1 (further only as CCL2) levels. It responded well to antidepressant treatment, rising to similar levels found in healthy controls. The authors hypothesized that CCL2 might act as a neuroprotective chemokine, positively affecting the central dopaminergic pathways [146]. Moreover, Janelidze et al., found that CCL2 levels in the plasma and CSF were significantly decreased in suicide attempters [147]. On the other hand, various contradictory results have been published correlating the increased serum levels of CCL2 with MDD development. For example, Simon et al., examined the serum level of 20 cytokines in total, and CCL2 was among the many elevated in MDD patients [148]. Although using a different methodology, Zhou et al., found a similar association between high plasma levels of CCL2 and the development of early pregnancy depression. The authors correlated its plasma levels with the levels of LPS as a marker of microbial translocation (the occurrence of gut microorganisms in extraintestinal locations) [149]. These conflicting reports indicate that the roles of CCL2 are pleiotropic, so the definitive interpretation of its central and peripheral levels in MDD patients has to be backed up by a more robust body of data. As reviewed by Curzytek and Leśkiewicz, the chemotactic activity of the CCL2–CCR2 axis (receptor–ligand axis) is only a part of the picture. It might as likely contribute to neurodegeneration and neuroinflammation as to neuroregeneration and neurotransmission [150].

16. CCL3

Macrophage inflammatory protein-1 alpha (MIP-1α or CCL3) is monokine encoded by the CCL3 gene, located at 17q12. It plays a crucial role during acute inflammation and is responsible for the recruitment and activation of polymorphonuclear leukocytes [151]. Hoge et al., showed that increased levels of MIP-1α with other pro-inflammatory cytokines are associated with panic disorder, post-traumatic stress disorder, and depression [152]. Merendino et al., demonstrated overexpressed MIP-1α in combination with fractalkine in patients with moderate to severe depression [153]. Depression with anxious distress is a clinically relevant subtype of MDD associated with higher levels of lipopolysaccharide-stimulated inflammatory markers, including MIP-1α [154]. Another study revealed a correlation between daytime melatonin with MIP-1α, CCL2, and VEGF-A in young patients with anxiety disorder [155]. A more recent study demonstrated that elevated serum levels of MIP-1α and iNOS are associated with post-stroke depression [156]. Moreover, increased levels of MIP-1α and other chemokines correlated with anxiety and depression during the late stages of pregnancy [157].

17. CCL5

The chemokine CCL5 (RANTES) is a chemokine encoded by the CCL5 gene with a cytogenetic location of 17q12. Data on CCL5 are scarce, as studies on this individual chemokine are only lately being conducted. CCL5, CCL2, and CCL11 were significantly associated with anxiety and depression in a study on the uterine–chemokine–brain axis [158]. A study on job stress outcomes reported lower serum CCL5 to correlate with higher anxiety scores [159]. Period Circadian Regulator 2 (Per2)-deficient mice were immune to centrally administered LPS upregulation of CCL5 and thus less likely to suffer from neuroinflammation-induced depression-like behavior [160]. A study on pregnant women in the third trimester correlated CCL5 and other chemokines with higher anxiety and anxiety/depression prevalence. However, after adjustment for clinical measures, the association with CCL5 did not prove significant [157]. A study from Polish authors reported higher CCL5 levels in subjects compared to controls, while the levels of IL-1β and IL-6 were significantly higher in subjects [161]. A meta-analysis of 73 studies did not find any significance in blood CCL5, while lower CCL4 and higher CCL-2,3,11 with CXCL7 and IL-8 were associated with depression [162].

18. CCL11

The CCL11 gene codes CCL11, also known as Eotaxin-1, with a cytogenetic location of 17q12. CCL11 is produced by T and B cells, endothelial cells, microglia, astrocytes, macrophages, eosinophils, and other cells in reaction to interaction with IL-4, IL-10, and IL-13. CCL11 functions also include a powerful attraction of eosinophils, basophils, neutrophils, macrophages, Th2 cells, and mastocyte cell precursors. In humans, its levels increase with age, and CCL11 is associated with cognitive and memory impairments [163][164][165]. In mice, CCL11 was found to promote microglia migration and activation with subsequent production of reactive oxygen species, potentiating glutamate-induced neuronal death [165]. In agreement with these findings, CCL11 levels in patients with bipolar disorder were found to negatively correlate with the left superior temporal volume, pointing to potential CLL-11 involvement in the pathophysiology of the disease via accelerated brain aging, and elevated CCL11 levels were also observed in MDD, regardless of the treatment [166][167]. Grassi-Oliveira et al. compared chemokine profiles in female patients with MDD with and without suicidal ideations and healthy controls. Although in their work, CCL11 was not elevated in suicidal compared to non-suicidal MDD patients, their findings confirm elevated CCL11 levels in MDD subjects compared to healthy controls [168]. However, later research comparing cytokine and chemokine profiles in depression and dysthymia reported elevated CCL11 in connection with other cytokines creating a specific network architecture in dysthymia.

19. Tumor Necrosis Factor

Tumor necrosis factor (TNF) is a multifunctional pro-inflammatory cytokine of the TNF superfamily encoded by the TNF gene located at 6p21.33 [169]. First introduced as TNF due to its capability of inducing necrosis in various neoplasms in mice, it was later renamed TNF-α because a previously described factor known as lymphotoxin was found to be homologous to TNF. Thus, its name was changed to TNF-β. However, the later discovery of a similar factor and its designation as lymphotoxin-β forced the researchers to change the name of TNF-β (former lymphotoxin) to lymphotoxin-α. Thus, the term TNF-β ceased to exist, and TNF-α became a meaningless “orphan term”. Therefore, in 1998, the official term of this cytokine was changed to its original TNF [170]. Despite that, a great bulk of studies out there still use the term TNF-α today.
A recent paper by Benedetti et al. evaluated the examination of peripheral levels of several pro-inflammatory cytokines, including the TNF, as a predictive method to assess the success of antidepressant therapy. TNF elevation at the baseline was associated with worse treatment outcomes [171]. Das et al., evaluated the relationship between TNF serum levels and MDD. They found that TNF was not only increased in MDD, but the levels were also directly proportional to its severity. Therefore, the peripheral levels of TNF might have a predictive value in clinical practice [172]. In 2020, Bialek et al., were the first to study the SNPs in various cytokine-coding genes, including the TNF gene, namely c.-1211T > C—TNF-α (rs1799964) and c.-488G > A—TNF-α (rs1800629) and its relation to MDD development and treatment effectiveness. The preliminary results showed that the C allele in the C/T genotype of rs179964 was associated with positive treatment outcomes and low serum levels of TNF. These results indicate that the molecular biological approach can provide additional information beneficial for a complex diagnostic assessment of MDD and its treatment prediction [173]. Ng et al., published a systematic review and meta-analysis on the relationship between peripheral levels of TNF and four other cytokines in the elderly diagnosed with depression and AD. The primary conclusion was that there was no difference in the TNF levels between study groups and controls in either of the reviewed disease entities [3]. On the contrary, a meta-analysis by Dowlati et al. found the opposite. Those studies that met the inclusion criteria showed a significant rise in TNF serum levels compared to controls [134]. All in all, peripheral TNF levels seem to reliably correlate with MDD development, its severity, and its response to treatment. However, whether the elevation of this pro-inflammatory cytokine is in a direct causal relationship with MDD and other psychiatric disorders is still obscure. It can also be the case of reverse causality or residual confounding. Such ambiguity applies to most of the discussed cytokines. These questions were addressed in a bi-directional two-sample Mendelian randomization study by Perry et al. Still, as far as TNF is concerned, the authors did not obtain any conclusive results [174].

20. Soluble TNF Receptor 2

Soluble TNF receptor 2 (sTNFR2) is a cleaved extracellular domain of the TNFR2, also known as tumor necrosis factor receptor superfamily member 1B (TNFRSF1B) encoded by the TNFRSF1B gene located at 1p36.22 [175]. The cleavage of sTNFR2 is carried out by the enzyme TACE, releasing it into circulation [176]. In 2017, Bobińska et al. conducted a genetic analysis of TNFR2 along with TNFR1 and TNF to investigate whether their increased expression correlates with recurrent depressive disorder (rDD). The n = 89 study group displayed a significantly higher expression of all the genes, and their expression was also found to be inversely proportional to the performance of the patient’s cognitive faculties, including learning effectiveness, working memory, attention, and others [177]. Yamamori et al., chose to address the limitations of standard diagnostic tests for mood disorders, including MDD, by evaluating the diagnostic utility of the multi-assay biological diagnostic test. The test combined the examination of plasma levels of sTNFR2 along with two other markers. The correct classification rate of the diagnosis based on the elevated sTNFR2 plasma levels and the other two markers was 69.2%, with 62.5% sensitivity and 82.5% specificity, suggesting the potential future diagnostic value of such an approach [178]. Papakostas et al. opted for a similar study design. Their multi-assay serum-based biological diagnostic test comprised nine markers, one of which was sTNFR2. The examination of this panel of markers, which included the elevated sTNFR2 levels, was 91.1% sensitive and 81.3% specific in correctly diagnosing MDD in the test subjects. Unsurprisingly, broadening the assay and appropriate marker selection has more significant benefits compared to a narrower panel. Still, overall, the diagnostic potential of muti-assay biological tests is high, addressing the downsides of standard tests [179]. Grassi-Oliveira et al. and Diniz et al. also reported increased serum/plasma levels of sTNFR2 in MDD patients and their potential value in clinical practice [180][181].
On the other hand, Schmidt et al. found sTNFR2 serum levels to be decreased in MDD. Based on these results, the authors hypothesized about the potential explanation of such a finding. The authors’ main interpretative avenue was that the sTNFR2 is essential in maintaining a normal balance of T regulatory (Treg) and T effector cells. Therefore, the absence or subpar activity of sTNFR2 may lead to T-cell differentiation modes associated with MDD. Another explanation might be that sTNFR2 has a vital role in the normal activity of several cortical regions associated with emotional responses such as the anterior cingulate cortex, dorsolateral prefrontal cortex, and hippocampus, so its under-expression might contribute to MDD pathogenesis [182]. This vital role of TNFR2 in normal brain function was also evidenced by Pillai et al., who measured CSF levels of sTNFR2 and evaluated two SNPs in the TNFRSF1B gene in patients with AD. The authors found that CSF levels of sTNFR2 and the specific TNFRSF1B gene variations can serve as markers of resilience to cognitive impairment associated with the disease [183]. Taken together, sTNFR2 seems to be yet another valuable analyte with the potential to enrich the modern diagnostic assessment of MDD patients, provided that its measurement is appropriately combined with other biomarkers.

21. Transforming Growth Factor-Beta

Transforming growth factor-beta (TGF-β) is a multifunctional cytokine that belongs to the TGF-β superfamily of soluble, dimeric peptides. Their primary function is to control and initiate the differentiation and proliferation of various types of cells. The TGFB1 gene encodes TGF-β with a cytogenetic location of 19q13.2. Signaling via the TGF-β/Smad pathway is essential in regulating stress response and has also been implicated in developing mood disorders [184]. TGF-β signaling has been researched concerning prenatal maternal depression and children’s brain development in utero [185]. Interestingly, lower gene expression of TGF-β was linked to larger amygdala volumes in these children. Deficits in signaling are not only common in patients with depression but also in AD [186]. In AD patients, targeting the TGF-β pathway could be a novel therapeutic approach [187]. In the inflammatory pathway, TGF-β acts as an anti-inflammatory factor, often countering the effects of IL-6. Their imbalance, combined with an imbalance in Th17/Treg cells, leads to chronic stress-induced depression in mice [188]. Moreover, gene polymorphisms in the TGFB gene (TT, +869) lead to lower TGF-β1 expression has been associated with impaired immunosuppression by Th3 cells and leading to depression [189]. A gene–environment (GxE) interaction study showed significant TGF-β and A2M (Alpha-2-Macroglobulin) interactions with emotional, physical, and sexual abuse [190]. Chronic fatigue syndrome in adolescents has been researched in a clinical trial that observed no significant differences in plasma samples, while TGF-β isoforms showed a relation to fatigue score [191]. All attributes mentioned above of TGF-β lead to the conclusion of a significant relation in the inflammatory depression pathway.

References

  1. Hart, B.L. Biological Basis of the Behavior of Sick Animals. Neurosci. Biobehav. Rev. 1988, 12, 123–137.
  2. Brebner, K. Synergistic Effects of Interleukin-1β, Interleukin-6, and Tumor Necrosis Factor-α Central Monoamine, Corticosterone, and Behavioral Variations. Neuropsychopharmacology 2000, 22, 566–580.
  3. Ng, A.; Tam, W.W.; Zhang, M.W.; Ho, C.S.; Husain, S.F.; McIntyre, R.S.; Ho, R.C. IL-1β, IL-6, TNF- α and CRP in Elderly Patients with Depression or Alzheimer’s Disease: Systematic Review and Meta-Analysis. Sci. Rep. 2018, 8, 12050.
  4. Ellul, P.; Boyer, L.; Groc, L.; Leboyer, M.; Fond, G. Interleukin-1 β-Targeted Treatment Strategies in Inflammatory Depression: Toward Personalized Care. Acta Psychiatr. Scand. 2016, 134, 469–484.
  5. Johnson, J.D.; Barnard, D.F.; Kulp, A.C.; Mehta, D.M. Neuroendocrine Regulation of Brain Cytokines After Psychological Stress. J. Endocr. Soc. 2019, 3, 1302–1320.
  6. Fleshner, M.; Crane, C.R. Exosomes, DAMPs and MiRNA: Features of Stress Physiology and Immune Homeostasis. Trends Immunol. 2017, 38, 768–776.
  7. Nguyen, K.T.; Deak, T.; Owens, S.M.; Kohno, T.; Fleshner, M.; Watkins, L.R.; Maier, S.F. Exposure to Acute Stress Induces Brain Interleukin-1β Protein in the Rat. J. Neurosci. 1998, 18, 2239–2246.
  8. Levine, J.; Barak, Y.; Chengappa, K.N.R.; Rapoport, A.; Rebey, M.; Barak, V. Cerebrospinal Cytokine Levels in Patients with Acute Depression. Neuropsychobiology 1999, 40, 171–176.
  9. Thomas, A.J.; Davis, S.; Morris, C.; Jackson, E.; Harrison, R.; O’Brien, J.T. Increase in Interleukin-1β in Late-Life Depression. AJP 2005, 162, 175–177.
  10. Corwin, E.J.; Johnston, N.; Pugh, L. Symptoms of Postpartum Depression Associated with Elevated Levels of Interleukin-1 Beta During the First Month Postpartum. Biol. Res. Nurs. 2008, 10, 128–133.
  11. Ferreira, A.M.; Leal, B.; Ferreira, I.; Brás, S.; Moreira, I.; Samões, R.; Sousa, A.P.; Santos, E.; Silva, B.; Costa, P.P.; et al. Depression and Anxiety in Multiple Sclerosis Patients: The Role of Genetic Variability of Interleukin 1β. Mult. Scler. Relat. Disord. 2021, 52, 102982.
  12. Ovaskainen, Y.; Koponen, H.; Jokelainen, J.; Keinänen-Kiukaanniemi, S.; Kumpusalo, E.; Vanhala, M. Depressive Symptomatology Is Associated with Decreased Interleukin-1 Beta and Increased Interleukin-1 Receptor Antagonist Levels in Males. Psychiatry Res. 2009, 167, 73–79.
  13. Seil, M.; Ouaaliti, M.E.; Abdou Foumekoye, S.; Pochet, S.; Dehaye, J. Distinct Regulation by Lipopolysaccharides of the Expression of Interleukin-1β by Murine Macrophages and Salivary Glands. Innate Immun. 2012, 18, 14–24.
  14. Clark, A.K.; Staniland, A.A.; Marchand, F.; Kaan, T.K.Y.; McMahon, S.B.; Malcangio, M. P2X7-Dependent Release of Interleukin-1 and Nociception in the Spinal Cord Following Lipopolysaccharide. J. Neurosci. 2010, 30, 573–582.
  15. Porterfield, V.M.; Gabella, K.M.; Simmons, M.A.; Johnson, J.D. Repeated Stressor Exposure Regionally Enhances Beta-Adrenergic Receptor-Mediated Brain IL-1β Production. Brain Behav. Immun. 2012, 26, 1249–1255.
  16. Barnard, D.F.; Gabella, K.M.; Kulp, A.C.; Parker, A.D.; Dugan, P.B.; Johnson, J.D. Sex Differences in the Regulation of Brain IL-1β in Response to Chronic Stress. Psychoneuroendocrinology 2019, 103, 203–211.
  17. Barrientos, R.M.; Sprunger, D.B.; Campeau, S.; Higgins, E.A.; Watkins, L.R.; Rudy, J.W.; Maier, S.F. Brain-Derived Neurotrophic Factor MRNA Downregulation Produced by Social Isolation Is Blocked by Intrahippocampal Interleukin-1 Receptor Antagonist. Neuroscience 2003, 121, 847–853.
  18. Schmid, A.W.; Lynch, M.A.; Herron, C.E. The Effects of IL-1 Receptor Antagonist on Beta Amyloid Mediated Depression of LTP in the Rat CA1 in Vivo. Hippocampus 2009, 19, 670–676.
  19. Wakabayashi, C.; Numakawa, T.; Odaka, H.; Ooshima, Y.; Kiyama, Y.; Manabe, T.; Kunugi, H.; Iwakura, Y. IL-1 Receptor-Antagonist (IL-1Ra) Knockout Mice Show Anxiety-like Behavior by Aging. Neurosci. Lett. 2015, 599, 20–25.
  20. Lombardo, S.D.; Mazzon, E.; Basile, M.S.; Cavalli, E.; Bramanti, P.; Nania, R.; Fagone, P.; Nicoletti, F.; Petralia, M.C. Upregulation of IL-1 Receptor Antagonist in a Mouse Model of Migraine. Brain Sci. 2019, 9, 172.
  21. Euteneuer, F.; Schwarz, M.J.; Dannehl, K.; Hartung, A.; Westermann, S.; Rief, W. Increased Soluble Interleukin-2 Receptor Levels Are Related to Somatic but Not to Cognitive-Affective Features in Major Depression. Brain Behav. Immun. 2012, 26, 1244–1248.
  22. Damoiseaux, J. The IL-2-IL-2 Receptor Pathway in Health and Disease: The Role of the Soluble IL-2 Receptor. Clin. Immunol. 2020, 218, 108515.
  23. Maes, M.; Bosmans, E.; Suy, E.; Vandervorst, C.; DeJonckheere, C.; Raus, J. Depression-Related Disturbances in Mitogen-Induced Lymphocyte Responses and Interleukin-1 Beta and Soluble Interleukin-2 Receptor Production. Acta Psychiatr. Scand. 1991, 84, 379–386.
  24. Al-Hakeim, H.K.; Al-Rammahi, D.A.; Al-Dujaili, A.H. IL-6, IL-18, SIL-2R, and TNFα Proinflammatory Markers in Depression and Schizophrenia Patients Who Are Free of Overt Inflammation. J. Affect. Disord. 2015, 182, 106–114.
  25. Liu, Y.; Ho, R.C.-M.; Mak, A. Interleukin (IL)-6, Tumour Necrosis Factor Alpha (TNF-α) 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, 230–239.
  26. IL3 Interleukin 3 -Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/3562 (accessed on 18 December 2022).
  27. Mangi, M.H.; Newland, A.C. Interleukin-3: Promises and Perspectives. Hematology 1998, 3, 55–66.
  28. Xiu, M.H.; Lin, C.G.; Tian, L.; Tan, Y.L.; Chen, J.; Chen, S.; Tan, S.P.; Wang, Z.R.; Yang, F.D.; Chen, D.C.; et al. Increased IL-3 Serum Levels in Chronic Patients with Schizophrenia: Associated with Psychopathology. Psychiatry Res. 2015, 229, 225–229.
  29. Gao, W.; Xu, Y.; Liang, J.; Sun, Y.; Zhang, Y.; Shan, F.; Ge, J.; Xia, Q. Comparison of Serum Cytokines Levels in Normal-Weight and Overweight Patients with First-Episode Drug-Naïve Major Depressive Disorder. Front. Endocrinol. (Lausanne) 2022, 13, 1048337.
  30. Osimo, E.F.; Pillinger, T.; Rodriguez, I.M.; Khandaker, G.M.; Pariante, C.M.; Howes, O.D. Inflammatory Markers in Depression: A Meta-Analysis of Mean Differences and Variability in 5,166 Patients and 5,083 Controls. Brain Behav. Immun. 2020, 87, 901–909.
  31. Qurashi, T.A.; Bhat, G.A.; Khan, M.S.; Rasool, R.; Sameen, F.; Hassan, I.; Mudassar, S. Interleukin 4 and Interleukin 4 Receptor Alpha Gene Variants and Risk of Atopy-A Case Control Study Based Assessment. Clin. Immunol. 2021, 229, 108783.
  32. Li, J.; Leschka, S.; Rutschow, S.; Schwimmbeck, P.L.; Husmann, L.; Noutsias, M.; Westermann, D.; Poller, W.; Zeichhardt, H.; Klingel, K.; et al. Immunomodulation by Interleukin-4 Suppresses Matrix Metalloproteinases and Improves Cardiac Function in Murine Myocarditis. Eur. J. Pharmacol. 2007, 554, 60–68.
  33. Quarta, A.; Berneman, Z.; Ponsaerts, P. Neuroprotective Modulation of Microglia Effector Functions Following Priming with Interleukin 4 and 13: Current Limitations in Understanding Their Mode-of-Action. Brain Behav. Immun. 2020, 88, 856–866.
  34. Zhang, J.; Rong, P.; Zhang, L.; He, H.; Zhou, T.; Fan, Y.; Mo, L.; Zhao, Q.; Han, Y.; Li, S.; et al. IL4-Driven Microglia Modulate Stress Resilience through BDNF-Dependent Neurogenesis. Sci. Adv. 2021, 7, eabb9888.
  35. Wachholz, S.; Knorr, A.; Mengert, L.; Plümper, J.; Sommer, R.; Juckel, G.; Friebe, A. Interleukin-4 Is a Participant in the Regulation of Depressive-like Behavior. Behav. Brain Res. 2017, 326, 165–172.
  36. Chou, Y.-H.; Hsieh, W.-C.; Chen, L.-C.; Lirng, J.-F.; Wang, S.-J. Association between the Serotonin Transporter and Cytokines: Implications for the Pathophysiology of Bipolar Disorder. J. Affect. Disord. 2016, 191, 29–35.
  37. Wachholz, S.; Eßlinger, M.; Manitz, M.-P.; Sommer, R.; Plümper, J.; Juckel, G.; Friebe, A. Interleukin-4−/− Mice Show a Depression-like Phenotype. Neurol. Psychiatry Brain Res. 2016, 22, 24.
  38. Schwenger, G.T.; Sanderson, C.J. New Directions in Understanding Interleukin-5 Gene Expression. Leuk. Lymphoma 1998, 28, 443–450.
  39. Elomaa, A.-P.; Niskanen, L.; Herzig, K.-H.; Viinamäki, H.; Hintikka, J.; Koivumaa-Honkanen, H.; Honkalampi, K.; Valkonen-Korhonen, M.; Harvima, I.T.; Lehto, S.M. Elevated Levels of Serum IL-5 Are Associated with an Increased Likelihood of Major Depressive Disorder. BMC Psychiatry 2012, 12, 2.
  40. Sawada, M.; Suzumura, A.; Itoh, Y.; Marunouchi, T. Production of Interleukin-5 by Mouse Astrocytes and Microglia in Culture. Neurosci. Lett. 1993, 155, 175–178.
  41. Ringheim, G.E. Mitogenic Effects of Interleukin-5 on Microglia. Neurosci. Lett. 1995, 201, 131–134.
  42. Köhler, C.A.; Freitas, T.H.; Maes, M.; de Andrade, N.Q.; Liu, C.S.; Fernandes, B.S.; Stubbs, B.; Solmi, M.; Veronese, N.; Herrmann, N.; et al. Peripheral Cytokine and Chemokine Alterations in Depression: A Meta-Analysis of 82 Studies. Acta Psychiatr. Scand. 2017, 135, 373–387.
  43. Ho, H.-Y.; Chin-Hung Chen, V.; Tzang, B.-S.; Hsieh, C.-C.; Wang, W.-K.; Weng, Y.-P.; Hsu, Y.-T.; Hsaio, H.-P.; Weng, J.-C.; Chen, Y.-L. Circulating Cytokines as Predictors of Depression in Patients with Breast Cancer. J. Psychiatr. Res. 2021, 136, 306–311.
  44. Hao, Y.; Zhou, Q.; Sun, Y.; Niu, W.; Du, J. Association of Three Single Nucleotide Polymorphisms in Interleukin 6 Gene with Risk of Chronic Obstructive Pulmonary Disease. Gene 2022, 828, 146467.
  45. Zadka, Ł.; Dzięgiel, P.; Kulus, M.; Olajossy, M. Clinical Phenotype of Depression Affects Interleukin-6 Synthesis. J. Interferon Cytokine Res. 2017, 37, 231–245.
  46. Li, Y.; Xie, Y.; Xu, Y.; Xian, X.; Wang, R.; Cai, L.; Li, G.; Li, Y. Interleukin-6-White Matter Network Differences Explained the Susceptibility to Depression after Stressful Life Events. J. Affect. Disord. 2022, 305, 122–132.
  47. Hodes, G.E.; Ménard, C.; Russo, S.J. Integrating Interleukin-6 into Depression Diagnosis and Treatment. Neurobiol. Stress 2016, 4, 15–22.
  48. Manfro, P.H.; Anselmi, L.; Barros, F.; Gonçalves, H.; Murray, J.; Oliveira, I.O.; Tovo-Rodrigues, L.; Wehrmeister, F.C.; Menezes, A.M.B.; Mondelli, V.; et al. Youth Depression and Inflammation: Cross-Sectional Network Analyses of C-Reactive Protein, Interleukin-6 and Symptoms in a Population-Based Sample. J. Psychiatr. Res. 2022, 150, 197–201.
  49. Young, J.J.; Bruno, D.; Pomara, N. A Review of the Relationship between Proinflammatory Cytokines and Major Depressive Disorder. J. Affect. Disord. 2014, 169, 15–20.
  50. Wang, P.; Feng, Y.; Wang, L.; Li, Y.; Fan, C.; Song, Q.; Yu, S.Y. Interleukin-6: Its Role and Mechanisms in Rescuing Depression-like Behaviors in Rat Models of Depression. Brain Behav. Immun. 2019, 82, 106–121.
  51. Schumertl, T.; Lokau, J.; Rose-John, S.; Garbers, C. Function and Proteolytic Generation of the Soluble Interleukin-6 Receptor in Health and Disease. Biochim. Biophys. Acta Mol. Cell Res. 2022, 1869, 119143.
  52. García-Juárez, M.; Camacho-Morales, A. Defining the Role of Anti- and Pro-Inflammatory Outcomes of Interleukin-6 in Mental Health. Neuroscience 2022, 492, 32–46.
  53. Ushiroyama, T.; Ikeda, A.; Ueki, M. Elevated Plasma Interleukin-6 (IL-6) and Soluble IL-6 Receptor Concentrations in Menopausal Women with and without Depression. Int. J. Gynaecol. Obs. 2002, 79, 51–52.
  54. Kelly, K.M.; Smith, J.A.; Mezuk, B. Depression and Interleukin-6 Signaling: A Mendelian Randomization Study. Brain Behav. Immun. 2021, 95, 106–114.
  55. Maes, M.; Meltzer, H.Y.; Bosmans, E.; Bergmans, R.; Vandoolaeghe, E.; Ranjan, R.; Desnyder, R. Increased Plasma Concentrations of Interleukin-6, Soluble Interleukin-6, Soluble Interleukin-2 and Transferrin Receptor in Major Depression. J. Affect. Disord. 1995, 34, 301–309.
  56. Tsai, S.-J. Role of Interleukin 8 in Depression and Other Psychiatric Disorders. Prog. Neuropsychopharmacol. Biol. Psychiatry 2021, 106, 110173.
  57. Kruse, J.L.; Olmstead, R.; Hellemann, G.; Breen, E.C.; Tye, S.J.; Brooks, J.O.; Wade, B.; Congdon, E.; Espinoza, R.; Narr, K.L.; et al. Interleukin-8 and Lower Severity of Depression in Females, but Not Males, with Treatment-Resistant Depression. J. Psychiatr. Res. 2021, 140, 350–356.
  58. Kruse, J.L.; Vasavada, M.M.; Olmstead, R.; Hellemann, G.; Wade, B.; Breen, E.C.; Brooks, J.O.; Congdon, E.; Espinoza, R.; Narr, K.L.; et al. Depression Treatment Response to Ketamine: Sex-Specific Role of Interleukin-8, but Not Other Inflammatory Markers. Transl. Psychiatry 2021, 11, 167.
  59. Kruse, J.L.; Olmstead, R.; Hellemann, G.; Wade, B.; Jiang, J.; Vasavada, M.M.; Brooks Iii, J.O.; Congdon, E.; Espinoza, R.; Narr, K.L.; et al. Inflammation and Depression Treatment Response to Electroconvulsive Therapy: Sex-Specific Role of Interleukin-8. Brain Behav. Immun. 2020, 89, 59–66.
  60. Irwin, M.R.; Olmstead, R.; Kruse, J.; Breen, E.C.; Haque, R. Association of Interleukin-8 and Risk of Incident and Recurrent Depression in Long-Term Breast Cancer Survivors. Brain Behav. Immun. 2022, 105, 131–138.
  61. Kern, S.; Skoog, I.; Börjesson-Hanson, A.; Blennow, K.; Zetterberg, H.; Ostling, S.; Kern, J.; Gudmundsson, P.; Marlow, T.; Rosengren, L.; et al. Higher CSF Interleukin-6 and CSF Interleukin-8 in Current Depression in Older Women. Results from a Population-Based Sample. Brain Behav. Immun. 2014, 41, 55–58.
  62. Kuzior, H.; Fiebich, B.L.; Yousif, N.M.; Saliba, S.W.; Ziegler, C.; Nickel, K.; Maier, S.J.; Süß, P.; Runge, K.; Matysik, M.; et al. Increased IL-8 Concentrations in the Cerebrospinal Fluid of Patients with Unipolar Depression. Compr. Psychiatry 2020, 102, 152196.
  63. Kruse, J.L.; Boyle, C.C.; Olmstead, R.; Breen, E.C.; Tye, S.J.; Eisenberger, N.I.; Irwin, M.R. Interleukin-8 and Depressive Responses to an Inflammatory Challenge: Secondary Analysis of a Randomized Controlled Trial. Sci. Rep. 2022, 12, 12627.
  64. Sajjad, M.U.; Blennow, K.; Knapskog, A.B.; Idland, A.-V.; Chaudhry, F.A.; Wyller, T.B.; Zetterberg, H.; Watne, L.O. Cerebrospinal Fluid Levels of Interleukin-8 in Delirium, Dementia, and Cognitively Healthy Patients. J. Alzheimers Dis. 2020, 73, 1363–1372.
  65. Chakraborty, S.; Kubatzky, K.F.; Mitra, D.K. An Update on Interleukin-9: From Its Cellular Source and Signal Transduction to Its Role in Immunopathogenesis. Int. J. Mol. Sci. 2019, 20, 2113.
  66. Donninelli, G.; Saraf-Sinik, I.; Mazziotti, V.; Capone, A.; Grasso, M.G.; Battistini, L.; Reynolds, R.; Magliozzi, R.; Volpe, E. Interleukin-9 Regulates Macrophage Activation in the Progressive Multiple Sclerosis Brain. J. Neuroinflamm. 2020, 17, 149.
  67. Shelton, R.C.; Claiborne, J.; Sidoryk-Wegrzynowicz, M.; Reddy, R.; Aschner, M.; Lewis, D.A.; Mirnics, K. Altered Expression of Genes Involved in Inflammation and Apoptosis in Frontal Cortex in Major Depression. Mol. Psychiatry 2011, 16, 751–762.
  68. Karlsson, L.; Nousiainen, N.; Scheinin, N.M.; Maksimow, M.; Salmi, M.; Lehto, S.M.; Tolvanen, M.; Lukkarinen, H.; Karlsson, H. Cytokine Profile and Maternal Depression and Anxiety Symptoms in Mid-Pregnancy—The FinnBrain Birth Cohort Study. Arch. Womens Ment. Health 2017, 20, 39–48.
  69. Varshney, P.; Parveen, R.; Khan, M.A.; Kohli, S.; Agarwal, N.B. Increased Serum Interleukin-9 and Interleukin-1β Are Associated with Depression in Type 2 Diabetes Patients. Arq. Neuro-Psiquiatr. 2020, 78, 255–261.
  70. Yui, S.; Sasayama, D.; Yamaguchi, M.; Washizuka, S. Altered Levels of Salivary Cytokines in Patients with Major Depressive Disorder. Clin. Neurol. Neurosurg. 2022, 221, 107390.
  71. Saraiva, M.; O’Garra, A. The Regulation of IL-10 Production by Immune Cells. Nat. Rev. Immunol. 2010, 10, 170–181.
  72. Yadav, U.; Kumar, P.; Rai, V. Interleukin-10 (IL-10) Gene Polymorphisms and Prostate Cancer Susceptibility: Evidence from a Meta-Analysis. Gene Rep. 2021, 25, 101377.
  73. Chi, C.-H.; Huang, Y.-Y.; Ye, S.-Z.; Shao, M.-M.; Jiang, M.-X.; Yang, M.-Y.; Wu, Q.; Shao, B.; Li, X.-M. Interleukin-10 Level Is Associated with Post-Stroke Depression in Acute Ischaemic Stroke Patients. J. Affect. Disord. 2021, 293, 254–260.
  74. Wiener, C.D.; Moreira, F.P.; Portela, L.V.; Strogulski, N.R.; Lara, D.R.; da Silva, R.A.; Souza, L.D.d.M.; Jansen, K.; Oses, J.P. Interleukin-6 and Interleukin-10 in Mood Disorders: A Population-Based Study. Psychiatry Res. 2019, 273, 685–689.
  75. Al-Fadhel, S.Z.; Al-Hakeim, H.K.; Al-Dujaili, A.H.; Maes, M. IL-10 Is Associated with Increased Mu-Opioid Receptor Levels in Major Depressive Disorder. Eur. Psychiatry 2019, 57, 46–51.
  76. Roque, S.; Correia-Neves, M.; Mesquita, A.R.; Palha, J.A.; Sousa, N. Interleukin-10: A Key Cytokine in Depression? Cardiovasc. Psychiatry Neurol. 2009, 2009, 187894.
  77. Kubera, M.; Maes, M.; Holan, V.; Basta-Kaim, A.; Roman, A.; Shani, J. Prolonged Desipramine Treatment Increases the Production of Interleukin-10, an Anti-Inflammatory Cytokine, in C57BL/6 Mice Subjected to the Chronic Mild Stress Model of Depression. J. Affect. Disord. 2001, 63, 171–178.
  78. Zhang, H.-Y.; Wang, Y.; He, Y.; Wang, T.; Huang, X.-H.; Zhao, C.-M.; Zhang, L.; Li, S.-W.; Wang, C.; Qu, Y.-N.; et al. A1 Astrocytes Contribute to Murine Depression-like Behavior and Cognitive Dysfunction, Which Can Be Alleviated by IL-10 or Fluorocitrate Treatment. J. Neuroinflamm. 2020, 17, 200.
  79. IL12A Interleukin 12A -Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/3592 (accessed on 30 November 2022).
  80. IL12B Interleukin 12B -Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/3593 (accessed on 30 November 2022).
  81. Liu, J.; Cao, S.; Kim, S.; Chung, E.Y.; Homma, Y.; Guan, X.; Jimenez, V.; Ma, X. Interleukin-12: An Update on Its Immunological Activities, Signaling and Regulation of Gene Expression. Curr. Immunol. Rev. 2005, 1, 119–137.
  82. Quinones, M.; Ahuja, S.K.; Melby, P.C.; Pate, L.; Reddick, R.L.; Ahuja, S.S. Preformed Membrane-Associated Stores of Interleukin (IL)-12 Are a Previously Unrecognized Source of Bioactive IL-12 That Is Mobilized within Minutes of Contact with an Intracellular Parasite. J. Exp. Med. 2000, 192, 507–516.
  83. Peluso, I.; Pallone, F.; Monteleone, G. Interleukin-12 and Th1 Immune Response in Crohn’s Disease: Pathogenetic Relevance and Therapeutic Implication. World J. Gastroenterol. 2006, 12, 5606–5610.
  84. Singh, V.K. Plasma Increase of Interleukin-12 and Interferon-Gamma. Pathological Significance in Autism. J. Neuroimmunol. 1996, 66, 143–145.
  85. Kim, Y.-K.; Suh, I.-B.; Kim, H.; Han, C.-S.; Lim, C.-S.; Choi, S.-H.; Licinio, J. The Plasma Levels of Interleukin-12 in Schizophrenia, Major Depression, and Bipolar Mania: Effects of Psychotropic Drugs. Mol. Psychiatry 2002, 7, 1107–1114.
  86. Lee, K.-M.; Kim, Y.-K. The Role of IL-12 and TGF-Beta1 in the Pathophysiology of Major Depressive Disorder. Int. Immunopharmacol. 2006, 6, 1298–1304.
  87. Sutcigil, L.; Oktenli, C.; Musabak, U.; Bozkurt, A.; Cansever, A.; Uzun, O.; Sanisoglu, S.Y.; Yesilova, Z.; Ozmenler, N.; Ozsahin, A.; et al. Pro- and Anti-Inflammatory Cytokine Balance in Major Depression: Effect of Sertraline Therapy. Clin. Dev. Immunol. 2007, 2007, 76396.
  88. Nowak, W.; Grendas, L.N.; Sanmarco, L.M.; Estecho, I.G.; Arena, Á.R.; Eberhardt, N.; Rodante, D.E.; Aoki, M.P.; Daray, F.M.; Carrera Silva, E.A.; et al. Pro-Inflammatory Monocyte Profile in Patients with Major Depressive Disorder and Suicide Behaviour and How Ketamine Induces Anti-Inflammatory M2 Macrophages by NMDAR and MTOR. EBioMedicine 2019, 50, 290–305.
  89. El-Tantawy, A.M.; El-Sayed, A.E.; Kora, B.A.; Amin, R.T. Psychiatric Morbidity Associated with Some Cytokines (IL-1beta, IL-12, IL-18 and TNF-Alpha) among Rheumatoid Arthritis Patients. Egypt J. Immunol. 2008, 15, 1–11.
  90. Schmidt, F.M.; Lichtblau, N.; Minkwitz, J.; Chittka, T.; Thormann, J.; Kirkby, K.C.; Sander, C.; Mergl, R.; Faßhauer, M.; Stumvoll, M.; et al. Cytokine Levels in Depressed and Non-Depressed Subjects, and Masking Effects of Obesity. J. Psychiatr. Res. 2014, 55, 29–34.
  91. Hou, R.; Ye, G.; Liu, Y.; Chen, X.; Pan, M.; Zhu, F.; Fu, J.; Fu, T.; Liu, Q.; Gao, Z.; et al. Effects of SSRIs on Peripheral Inflammatory Cytokines in Patients with Generalized Anxiety Disorder. Brain Behav. Immun. 2019, 81, 105–110.
  92. Liu, J.J.; Wei, Y.B.; Strawbridge, R.; Bao, Y.; Chang, S.; Shi, L.; Que, J.; Gadad, B.S.; Trivedi, M.H.; Kelsoe, J.R.; et al. Peripheral Cytokine Levels and Response to Antidepressant Treatment in Depression: A Systematic Review and Meta-Analysis. Mol. Psychiatry 2020, 25, 339–350.
  93. IL13 Interleukin 13 -Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/3596 (accessed on 30 November 2022).
  94. Minty, A.; Chalon, P.; Derocq, J.M.; Dumont, X.; Guillemot, J.C.; Kaghad, M.; Labit, C.; Leplatois, P.; Liauzun, P.; Miloux, B. Interleukin-13 Is a New Human Lymphokine Regulating Inflammatory and Immune Responses. Nature 1993, 362, 248–250.
  95. Kolosowska, N.; Keuters, M.H.; Wojciechowski, S.; Keksa-Goldsteine, V.; Laine, M.; Malm, T.; Goldsteins, G.; Koistinaho, J.; Dhungana, H. Peripheral Administration of IL-13 Induces Anti-Inflammatory Microglial/Macrophage Responses and Provides Neuroprotection in Ischemic Stroke. Neurotherapeutics 2019, 16, 1304–1319.
  96. Khalil, S.M.; Bernstein, I.; Kulaga, H.; Gour, N.; Rowan, N.; Lajoie, S.; Lane, A.P. Interleukin 13 (IL-13) Alters Hypoxia-Associated Genes and Upregulates CD73. Int. Forum Allergy Rhinol. 2020, 10, 1096–1102.
  97. Doherty, T.M.; Kastelein, R.; Menon, S.; Andrade, S.; Coffman, R.L. Modulation of Murine Macrophage Function by IL-13. J. Immunol. 1993, 151, 7151–7160.
  98. Corren, J. Role of Interleukin-13 in Asthma. Curr. Allergy Asthma Rep. 2013, 13, 415–420.
  99. Iwaszko, M.; Biały, S.; Bogunia-Kubik, K. Significance of Interleukin (IL)-4 and IL-13 in Inflammatory Arthritis. Cells 2021, 10, 3000.
  100. Pavón, L.; Sandoval-López, G.; Eugenia Hernández, M.; Loría, F.; Estrada, I.; Pérez, M.; Moreno, J.; Avila, U.; Leff, P.; Antón, B.; et al. Th2 Cytokine Response in Major Depressive Disorder Patients before Treatment. J. Neuroimmunol. 2006, 172, 156–165.
  101. Matalka, K.Z. Neuroendocrine and Cytokines-Induced Responses to Minutes, Hours, and Days of Mental Stress. Neuro Endocrinol. Lett. 2003, 24, 283–292.
  102. Hernández, M.E.; Mendieta, D.; Martínez-Fong, D.; Loría, F.; Moreno, J.; Estrada, I.; Bojalil, R.; Pavón, L. Variations in Circulating Cytokine Levels during 52 Week Course of Treatment with SSRI for Major Depressive Disorder. Eur. Neuropsychopharmacol. 2008, 18, 917–924.
  103. Wong, M.-L.; Dong, C.; Maestre-Mesa, J.; Licinio, J. Polymorphisms in Inflammation-Related Genes Are Associated with Susceptibility to Major Depression and Antidepressant Response. Mol. Psychiatry 2008, 13, 800–812.
  104. Czysz, A.H.; Mason, B.L.; Li, Q.; Chin-Fatt, C.; Minhajuddin, A.; Carmody, T.; Trivedi, M.H. Comparison of Inflammatory Markers as Moderators of Depression Outcomes: A CO-MED Study. J. Affect. Disord. 2021, 295, 1066–1071.
  105. Siwek, M.; Sowa-Kućma, M.; Dudek, D.; Styczeń, K.; Szewczyk, B.; Kotarska, K.; Misztak, P.; Pilc, A.; Wolak, M.; Nowak, G. Oxidative Stress Markers in Affective Disorders. Pharmacol. Rep. 2013, 65, 1558–1571.
  106. Vai, B.; Mazza, M.G.; Cazzetta, S.; Calesella, F.; Aggio, V.; Lorenzi, C.; Zanardi, R.; Poletti, S.; Colombo, C.; Benedetti, F. Higher Interleukin 13 Differentiates Patients with a Positive History of Suicide Attempts in Major Depressive Disorder. J. Affect. Disord. Rep. 2021, 6, 100254.
  107. Timonen, M.; Jokelainen, J.; Hakko, H.; Silvennoinen-Kassinen, S.; Meyer-Rochow, V.B.; Herva, A.; Räsänen, P. Atopy and Depression: Results from the Northern Finland 1966 Birth Cohort Study. Mol. Psychiatry 2003, 8, 738–744.
  108. Timonen, M.; Viilo, K.; Hakko, H.; Särkioja, T.; Meyer-Rochow, V.B.; Väisänen, E.; Räsänen, P. Is Seasonality of Suicides Stronger in Victims with Hospital-Treated Atopic Disorders? Psychiatry Res. 2004, 126, 167–175.
  109. Tonelli, L.H.; Stiller, J.; Rujescu, D.; Giegling, I.; Schneider, B.; Maurer, K.; Schnabel, A.; Möller, H.-J.; Chen, H.H.; Postolache, T.T. Elevated Cytokine Expression in the Orbitofrontal Cortex of Victims of Suicide. Acta Psychiatr. Scand. 2008, 117, 198–206.
  110. IL17A Interleukin 17A -Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/3605 (accessed on 13 December 2022).
  111. Rouvier, E.; Luciani, M.F.; Mattéi, M.G.; Denizot, F.; Golstein, P. CTLA-8, Cloned from an Activated T Cell, Bearing AU-Rich Messenger RNA Instability Sequences, and Homologous to a Herpesvirus Saimiri Gene. J. Immunol. 1993, 150, 5445–5456.
  112. McGeachy, M.J.; Cua, D.J.; Gaffen, S.L. The IL-17 Family of Cytokines in Health and Disease. Immunity 2019, 50, 892–906.
  113. Matusevicius, D.; Kivisäkk, P.; He, B.; Kostulas, N.; Ozenci, V.; Fredrikson, S.; Link, H. Interleukin-17 MRNA Expression in Blood and CSF Mononuclear Cells Is Augmented in Multiple Sclerosis. Mult. Scler. 1999, 5, 101–104.
  114. Nothdurfter, C.; Milenkovic, V.M.; Sarubin, N.; Hilbert, S.; Manook, A.; Weigl, J.; Almeqbaali, K.; Wetzel, C.H.; Rupprecht, R.; Baghai, T.C. The Cytokine IL-17A as a Marker of Treatment Resistance in Major Depressive Disorder? Eur. J. Neurosci. 2021, 53, 172–182.
  115. Medina-Rodriguez, E.M.; Madorma, D.; O’Connor, G.; Mason, B.L.; Han, D.; Deo, S.K.; Oppenheimer, M.; Nemeroff, C.B.; Trivedi, M.H.; Daunert, S.; et al. Identification of a Signaling Mechanism by Which the Microbiome Regulates Th17 Cell-Mediated Depressive-Like Behaviors in Mice. Am. J. Psychiatry 2020, 177, 974–990.
  116. Alvarez-Mon, M.A.; Gómez-Lahoz, A.M.; Orozco, A.; Lahera, G.; Diaz, D.; Ortega, M.A.; Albillos, A.; Quintero, J.; Aubá, E.; Monserrat, J.; et al. Expansion of CD4 T Lymphocytes Expressing Interleukin 17 and Tumor Necrosis Factor in Patients with Major Depressive Disorder. J. Pers. Med. 2021, 11, 220.
  117. Min, Z.; Li, Y.; Ying, H. Blood T-Helper 17 Cells and Interleukin-17A Correlate with the Elevated Risk of Postpartum Depression and Anxiety. J. Clin. Lab. Anal. 2022, 36, e24559.
  118. IL18 Interleukin 18 -Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/3606 (accessed on 30 November 2022).
  119. Nakamura, K.; Okamura, H.; Wada, M.; Nagata, K.; Tamura, T. Endotoxin-Induced Serum Factor That Stimulates Gamma Interferon Production. Infect. Immun. 1989, 57, 590–595.
  120. Felderhoff-Mueser, U.; Schmidt, O.I.; Oberholzer, A.; Bührer, C.; Stahel, P.F. IL-18: A Key Player in Neuroinflammation and Neurodegeneration? Trends Neurosci. 2005, 28, 487–493.
  121. Zheng, D.; Liwinski, T.; Elinav, E. Inflammasome Activation and Regulation: Toward a Better Understanding of Complex Mechanisms. Cell Discov. 2020, 6, 36.
  122. Wong, M.-L.; Inserra, A.; Lewis, M.D.; Mastronardi, C.A.; Leong, L.; Choo, J.; Kentish, S.; Xie, P.; Morrison, M.; Wesselingh, S.L.; et al. Inflammasome Signaling Affects Anxiety- and Depressive-like Behavior and Gut Microbiome Composition. Mol. Psychiatry 2016, 21, 797–805.
  123. Song, A.-Q.; Gao, B.; Fan, J.-J.; Zhu, Y.-J.; Zhou, J.; Wang, Y.-L.; Xu, L.-Z.; Wu, W.-N. NLRP1 Inflammasome Contributes to Chronic Stress-Induced Depressive-like Behaviors in Mice. J. Neuroinflamm. 2020, 17, 178.
  124. Fan, N.; Luo, Y.; Ou, Y.; He, H. Altered Serum Levels of TNF-α, IL-6, and IL-18 in Depressive Disorder Patients. Hum. Psychopharmacol. 2017, 32, e2588.
  125. Kokai, M.; Kashiwamura, S.; Okamura, H.; Ohara, K.; Morita, Y. Plasma Interleukin-18 Levels in Patients with Psychiatric Disorders. J. Immunother. 2002, 25 (Suppl. S1), S68–S71.
  126. Alcocer-Gómez, E.; de Miguel, M.; Casas-Barquero, N.; Núñez-Vasco, J.; Sánchez-Alcazar, J.A.; Fernández-Rodríguez, A.; Cordero, M.D. NLRP3 Inflammasome Is Activated in Mononuclear Blood Cells from Patients with Major Depressive Disorder. Brain Behav. Immun. 2014, 36, 111–117.
  127. Prossin, A.R.; Koch, A.E.; Campbell, P.L.; McInnis, M.G.; Zalcman, S.S.; Zubieta, J.-K. Association of Plasma Interleukin-18 Levels with Emotion Regulation and μ-Opioid Neurotransmitter Function in Major Depression and Healthy Volunteers. Biol. Psychiatry 2011, 69, 808–812.
  128. Witek-Janusek, L.; Albuquerque, K.; Chroniak, K.R.; Chroniak, C.; Durazo, R.; Mathews, H.L. Effect of Mindfulness Based Stress Reduction on Immune Function, Quality of Life and Coping In Women Newly Diagnosed with Early Stage Breast Cancer. Brain Behav. Immun. 2008, 22, 969–981.
  129. Andersen, B.L.; Farrar, W.B.; Golden-Kreutz, D.; Kutz, L.A.; MacCallum, R.; Courtney, M.E.; Glaser, R. Stress and Immune Responses after Surgical Treatment for Regional Breast Cancer. J. Natl. Cancer Inst. 1998, 90, 30–36.
  130. Andersen, B.L.; Farrar, W.B.; Golden-Kreutz, D.M.; Glaser, R.; Emery, C.F.; Crespin, T.R.; Shapiro, C.L.; Carson, W.E. Psychological, Behavioral, and Immune Changes after a Psychological Intervention: A Clinical Trial. J. Clin. Oncol. 2004, 22, 3570–3580.
  131. Maes, M.; Song, C.; Lin, A.-H.; Bonaccorso, S.; Kenis, G.; De Jongh, R.; Bosmans, E.; Scharpé, S. Negative Immunoregulatory Effects of Antidepressants: Inhibition of Interferon-γ and Stimulation of Interleukin-10 Secretion. Neuropsychopharmacology 1999, 20, 370–379.
  132. Pokryszko-Dragan, A.; Frydecka, I.; Kosmaczewska, A.; Ciszak, L.; Bilińska, M.; Gruszka, E.; Podemski, R.; Frydecka, D. Stimulated Peripheral Production of Interferon-Gamma Is Related to Fatigue and Depression in Multiple Sclerosis. Clin. Neurol. Neurosurg. 2012, 114, 1153–1158.
  133. Mandolesi, G.; Bullitta, S.; Fresegna, D.; Gentile, A.; De Vito, F.; Dolcetti, E.; Rizzo, F.R.; Strimpakos, G.; Centonze, D.; Musella, A. Interferon-γ Causes Mood Abnormalities by Altering Cannabinoid CB1 Receptor Function in the Mouse Striatum. Neurobiol. Dis. 2017, 108, 45–53.
  134. Dowlati, Y.; Herrmann, N.; Swardfager, W.; Liu, H.; Sham, L.; Reim, E.K.; Lanctôt, K.L. A Meta-Analysis of Cytokines in Major Depression. Biol. Psychiatry 2010, 67, 446–457.
  135. Daria, S.; Proma, M.A.; Shahriar, M.; Islam, S.M.A.; Bhuiyan, M.A.; Islam, M.R. Serum Interferon-Gamma Level Is Associated with Drug-Naïve Major Depressive Disorder. SAGE Open Med. 2020, 8, 2050312120974169.
  136. Maes, M.; Scharpé, S.; Meltzer, H.Y.; Okayli, G.; Bosmans, E.; D’Hondt, P.; Vanden Bossche, B.V.; Cosyns, P. Increased Neopterin and Interferon-Gamma Secretion and Lower Availability of L-Tryptophan in Major Depression: Further Evidence for an Immune Response. Psychiatry Res. 1994, 54, 143–160.
  137. Nam, J.-H.; Cha, B.; Park, J.-Y.; Abekura, F.; Kim, C.-H.; Kim, J.-R. Mitogen-Induced Interferon Gamma Production in Human Whole Blood: The Effect of Heat and Cations. Curr. Pharm. Biotechnol. 2019, 20, 562–572.
  138. Zhang, J.; He, H.; Qiao, Y.; Zhou, T.; He, H.; Yi, S.; Zhang, L.; Mo, L.; Li, Y.; Jiang, W.; et al. Priming of Microglia with IFN-γ Impairs Adult Hippocampal Neurogenesis and Leads to Depression-like Behaviors and Cognitive Defects. Glia 2020, 68, 2674–2692.
  139. Pusic, A.D.; Mitchell, H.M.; Kunkler, P.E.; Klauer, N.; Kraig, R.P. Spreading Depression Transiently Disrupts Myelin via Interferon-Gamma Signaling. Exp. Neurol. 2015, 264, 43–54.
  140. Oxenkrug, G.; Perianayagam, M.; Mikolich, D.; Requintina, P.; Shick, L.; Ruthazer, R.; Zucker, D.; Summergrad, P. Interferon-Gamma (+874) T/A Genotypes and Risk of IFN-Alpha-Induced Depression. J. Neural Transm. (Vienna) 2011, 118, 271–274.
  141. Mariani, N.; Everson, J.; Pariante, C.M.; Borsini, A. Modulation of Microglial Activation by Antidepressants. J. Psychopharmacol. 2022, 36, 131–150.
  142. Myint, A.M.; Bondy, B.; Baghai, T.C.; Eser, D.; Nothdurfter, C.; Schüle, C.; Zill, P.; Müller, N.; Rupprecht, R.; Schwarz, M.J. Tryptophan Metabolism and Immunogenetics in Major Depression: A Role for Interferon-γ Gene. Brain Behav. Immun. 2013, 31, 128–133.
  143. CCL2 C-C Motif Chemokine Ligand 2 -Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/6347 (accessed on 13 December 2022).
  144. Bose, S.; Cho, J. Role of Chemokine CCL2 and Its Receptor CCR2 in Neurodegenerative Diseases. Arch. Pharm. Res. 2013, 36, 1039–1050.
  145. Proma, M.A.; Daria, S.; Nahar, Z.; Ashraful Islam, S.M.; Bhuiyan, M.A.; Islam, M.R. Monocyte Chemoattractant Protein-1 Levels Are Associated with Major Depressive Disorder. J. Basic Clin. Physiol. Pharm. 2022, 33, 735–741.
  146. Myung, W.; Lim, S.-W.; Woo, H.I.; Park, J.H.; Shim, S.; Lee, S.-Y.; Kim, D.K. Serum Cytokine Levels in Major Depressive Disorder and Its Role in Antidepressant Response. Psychiatry Investig. 2016, 13, 644–651.
  147. Janelidze, S.; Ventorp, F.; Erhardt, S.; Hansson, O.; Minthon, L.; Flax, J.; Samuelsson, M.; Traskman-Bendz, L.; Brundin, L. Altered Chemokine Levels in the Cerebrospinal Fluid and Plasma of Suicide Attempters. Psychoneuroendocrinology 2013, 38, 853–862.
  148. Simon, N.M.; McNamara, K.; Chow, C.W.; Maser, R.S.; Papakostas, G.I.; Pollack, M.H.; Nierenberg, A.A.; Fava, M.; Wong, K.K. A Detailed Examination of Cytokine Abnormalities in Major Depressive Disorder. Eur. Neuropsychopharmacol. 2008, 18, 230–233.
  149. Zhou, Z.; Guille, C.; Ogunrinde, E.; Liu, R.; Luo, Z.; Powell, A.; Jiang, W. Increased Systemic Microbial Translocation Is Associated with Depression during Early Pregnancy. J. Psychiatr. Res. 2018, 97, 54–57.
  150. Curzytek, K.; Leśkiewicz, M. Targeting the CCL2-CCR2 Axis in Depressive Disorders. Pharm. Rep. 2021, 73, 1052–1062.
  151. Marciniak, E.; Faivre, E.; Dutar, P.; Alves Pires, C.; Demeyer, D.; Caillierez, R.; Laloux, C.; Buée, L.; Blum, D.; Humez, S. The Chemokine MIP-1α/CCL3 Impairs Mouse Hippocampal Synaptic Transmission, Plasticity and Memory. Sci. Rep. 2015, 5, 15862.
  152. Hoge, E.A.; Brandstetter, K.; Moshier, S.; Pollack, M.H.; Wong, K.K.; Simon, N.M. Broad Spectrum of Cytokine Abnormalities in Panic Disorder and Posttraumatic Stress Disorder. Depress. Anxiety 2009, 26, 447–455.
  153. Merendino, R.A.; Di Pasquale, G.; De Luca, F.; Di Pasquale, L.; Ferlazzo, E.; Martino, G.; Palumbo, M.C.; Morabito, F.; Gangemi, S. Involvement of Fractalkine and Macrophage Inflammatory Protein-1 Alpha in Moderate-Severe Depression. Mediat. Inflamm. 2004, 13, 205–207.
  154. Gaspersz, R.; Lamers, F.; Wittenberg, G.; Beekman, A.T.F.; van Hemert, A.M.; Schoevers, R.A.; Penninx, B.W.J.H. The Role of Anxious Distress in Immune Dysregulation in Patients with Major Depressive Disorder. Transl. Psychiatry 2017, 7, 1268.
  155. Sundberg, I.; Rasmusson, A.J.; Ramklint, M.; Just, D.; Ekselius, L.; Cunningham, J.L. Daytime Melatonin Levels in Saliva Are Associated with Inflammatory Markers and Anxiety Disorders. Psychoneuroendocrinology 2020, 112, 104514.
  156. Wang, X.; Fang, C.; Liu, X.; Wei, W.; Zhang, M.; Chen, S.; Shi, F. High Serum Levels of INOS and MIP-1α Are Associated with Post-Stroke Depression. Neuropsychiatr. Dis. Treat. 2021, 17, 2481–2487.
  157. Camacho-Arroyo, I.; Flores-Ramos, M.; Mancilla-Herrera, I.; Cruz, F.M.C.; Hernández-Ruiz, J.; Diaz, G.P.; Labonne, B.F.; Del Pilar Meza-Rodríguez, M.; Gelman, P.L. Chemokine Profile in Women with Moderate to Severe Anxiety and Depression during Pregnancy. BMC Pregnancy Childbirth 2021, 21, 807.
  158. Roomruangwong, C.; Sirivichayakul, S.; Carvalho, A.F.; Maes, M. The Uterine-Chemokine-Brain Axis: Menstrual Cycle-Associated Symptoms (MCAS) Are in Part Mediated by CCL2, CCL5, CCL11, CXCL8 and CXCL10. J. Affect. Disord. 2020, 269, 85–93.
  159. Polacchini, A.; Girardi, D.; Falco, A.; Zanotta, N.; Comar, M.; De Carlo, N.A.; Tongiorgi, E. Distinct CCL2, CCL5, CCL11, CCL27, IL-17, IL-6, BDNF Serum Profiles Correlate to Different Job-Stress Outcomes. Neurobiol. Stress 2018, 8, 82–91.
  160. Chen, X.; Hu, Q.; Zhang, K.; Teng, H.; Li, M.; Li, D.; Wang, J.; Du, Q.; Zhao, M. The Clock-Controlled Chemokine Contributes to Neuroinflammation-Induced Depression. FASEB J. 2020, 34, 8357–8366.
  161. Małujło-Balcerska, E.; Kumor-Kisielewska, A.; Szemraj, J.; Pietras, T. Chemokine (C-C Motif) Ligand 5 (RANTES) Concentrations in the Peripheral Blood of Patients with a Depressive Disorder. Pharm. Rep. 2022, 74, 759–768.
  162. Leighton, S.P.; Nerurkar, L.; Krishnadas, R.; Johnman, C.; Graham, G.J.; Cavanagh, J. Chemokines in Depression in Health and in Inflammatory Illness: A Systematic Review and Meta-Analysis. Mol. Psychiatry 2018, 23, 48–58.
  163. Ivanovska, M.; Abdi, Z.; Murdjeva, M.; Macedo, D.; Maes, A.; Maes, M. CCL-11 or Eotaxin-1: An Immune Marker for Ageing and Accelerated Ageing in Neuro-Psychiatric Disorders. Pharmaceuticals 2020, 13, 230.
  164. Máchal, J.; Vašků, A.; Kincl, V.; Hlavna, M.; Bartáková, V.; Jurajda, M.; Meluzín, J. Association between Three Single Nucleotide Polymorphisms in Eotaxin (CCL 11) Gene, Hexanucleotide Repetition Upstream, Severity and Course of Coronary Atherosclerosis. J. Appl. Genet. 2012, 53, 271–278.
  165. Teixeira, A.L.; Gama, C.S.; Rocha, N.P.; Teixeira, M.M. Revisiting the Role of Eotaxin-1/CCL11 in Psychiatric Disorders. Front. Psychiatry 2018, 9, 241.
  166. Mohite, S.; Cordeiro, T.; Tannous, J.; Mwangi, B.; Selvaraj, S.; Soares, J.C.; Sanches, M.; Teixeira, A.L. Eotaxin-1/CCL11 Correlates with Left Superior Temporal Gyrus in Bipolar Disorder: A Preliminary Report Suggesting Accelerated Brain Aging. J. Affect. Disord. 2020, 273, 592–596.
  167. Magalhaes, P.V.S.; Jansen, K.; Stertz, L.; Ferrari, P.; Pinheiro, R.T.; da Silva, R.A.; Kapczinski, F. Peripheral Eotaxin-1 (CCL11) Levels and Mood Disorder Diagnosis in a Population-Based Sample of Young Adults. J. Psychiatr. Res. 2014, 48, 13–15.
  168. Grassi-Oliveira, R.; Brieztke, E.; Teixeira, A.; Pezzi, J.C.; Zanini, M.; Lopes, R.P.; Bauer, M.E. Peripheral Chemokine Levels in Women with Recurrent Major Depression with Suicidal Ideation. Braz J. Psychiatry 2012, 34, 71–75.
  169. TNF Tumor Necrosis Factor -Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/7124 (accessed on 30 November 2022).
  170. Grimstad, Ø. Tumor Necrosis Factor and the Tenacious α. JAMA Derm. 2016, 152, 557.
  171. Benedetti, F.; Poletti, S.; Vai, B.; Mazza, M.G.; Lorenzi, C.; Brioschi, S.; Aggio, V.; Branchi, I.; Colombo, C.; Furlan, R.; et al. Higher Baseline Interleukin-1β and TNF-α Hamper Antidepressant Response in Major Depressive Disorder. Eur. Neuropsychopharmacol. 2021, 42, 35–44.
  172. Das, R.; Emon, M.P.Z.; Shahriar, M.; Nahar, Z.; Islam, S.M.A.; Bhuiyan, M.A.; Islam, S.N.; Islam, M.R. Higher Levels of Serum IL-1β and TNF-α Are Associated with an Increased Probability of Major Depressive Disorder. Psychiatry Res. 2021, 295, 113568.
  173. Bialek, K.; Czarny, P.; Watala, C.; Synowiec, E.; Wigner, P.; Bijak, M.; Talarowska, M.; Galecki, P.; Szemraj, J.; Sliwinski, T. Preliminary Study of the Impact of Single-Nucleotide Polymorphisms of IL-1α, IL-1β and TNF-α Genes on the Occurrence, Severity and Treatment Effectiveness of the Major Depressive Disorder. Cell. Mol. Neurobiol. 2020, 40, 1049–1056.
  174. Perry, B.I.; Upthegrove, R.; Kappelmann, N.; Jones, P.B.; Burgess, S.; Khandaker, G.M. Associations of Immunological Proteins/Traits with Schizophrenia, Major Depression and Bipolar Disorder: A Bi-Directional Two-Sample Mendelian Randomization Study. Brain Behav. Immun. 2021, 97, 176–185.
  175. TNFRSF1B TNF Receptor Superfamily Member 1B -Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/7133 (accessed on 30 November 2022).
  176. Black, R.A. Tumor Necrosis Factor-Alpha Converting Enzyme. Int. J. Biochem. Cell. Biol. 2002, 34, 1–5.
  177. Bobińska, K.; Gałecka, E.; Szemraj, J.; Gałecki, P.; Talarowska, M. Is There a Link between TNF Gene Expression and Cognitive Deficits in Depression? Acta Biochim. Pol. 2017, 64, 65–73.
  178. Yamamori, H.; Ishima, T.; Yasuda, Y.; Fujimoto, M.; Kudo, N.; Ohi, K.; Hashimoto, K.; Takeda, M.; Hashimoto, R. Assessment of a Multi-Assay Biological Diagnostic Test for Mood Disorders in a Japanese Population. Neurosci. Lett. 2016, 612, 167–171.
  179. Papakostas, G.I.; Shelton, R.C.; Kinrys, G.; Henry, M.E.; Bakow, B.R.; Lipkin, S.H.; Pi, B.; Thurmond, L.; Bilello, J.A. Assessment of a Multi-Assay, Serum-Based Biological Diagnostic Test for Major Depressive Disorder: A Pilot and Replication Study. Mol. Psychiatry 2013, 18, 332–339.
  180. Grassi-Oliveira, R.; Brietzke, E.; Pezzi, J.C.; Lopes, R.P.; Teixeira, A.L.; Bauer, M.E. Increased Soluble Tumor Necrosis Factor-Alpha Receptors in Patients with Major Depressive Disorder. Psychiatry Clin. Neurosci. 2009, 63, 202–208.
  181. Diniz, B.S.; Teixeira, A.L.; Talib, L.L.; Mendonça, V.A.; Gattaz, W.F.; Forlenza, O.V. Increased Soluble TNF Receptor 2 in Antidepressant-Free Patients with Late-Life Depression. J. Psychiatr. Res. 2010, 44, 917–920.
  182. Schmidt, F.M.; Koch, J.; Nowak, C.; Holdt, L.M.; Teupser, D.; Hegerl, U.; Himmerich, H. Ligands and Receptors of the TNF Superfamily Are Decreased in Major Depression and during Early Antidepressant Therapy. J. Psychiatr. Res. 2019, 119, 116–121.
  183. Pillai, J.A.; Bebek, G.; Khrestian, M.; Bena, J.; Bergmann, C.C.; Bush, W.S.; Leverenz, J.B.; Bekris, L.M. TNFRSF1B Gene Variants and Related Soluble TNFR2 Levels Impact Resilience in Alzheimer’s Disease. Front. Aging Neurosci. 2021, 13, 638922.
  184. Hiew, L.-F.; Poon, C.-H.; You, H.-Z.; Lim, L.-W. TGF-β/Smad Signalling in Neurogenesis: Implications for Neuropsychiatric Diseases. Cells 2021, 10, 1382.
  185. Qiu, A.; Zhang, H.; Wang, C.; Chong, Y.-S.; Shek, L.P.; Gluckman, P.D.; Meaney, M.J.; Fortier, M.V.; Wu, Y. Canonical TGF-β Signaling Regulates the Relationship between Prenatal Maternal Depression and Amygdala Development in Early Life. Transl. Psychiatry 2021, 11, 170.
  186. Caraci, F.; Spampinato, S.F.; Morgese, M.G.; Tascedda, F.; Salluzzo, M.G.; Giambirtone, M.C.; Caruso, G.; Munafò, A.; Torrisi, S.A.; Leggio, G.M.; et al. Neurobiological Links between Depression and AD: The Role of TGF-Β1 Signaling as a New Pharmacological Target. Pharm. Res. 2018, 130, 374–384.
  187. Grasso, M.; Caruso, G.; Godos, J.; Bonaccorso, A.; Carbone, C.; Castellano, S.; Currenti, W.; Grosso, G.; Musumeci, T.; Caraci, F. Improving Cognition with Nutraceuticals Targeting TGF-Β1 Signaling. Antioxidants 2021, 10, 1075.
  188. Huang, C.; Zhang, F.; Li, P.; Song, C. Low-Dose IL-2 Attenuated Depression-like Behaviors and Pathological Changes through Restoring the Balances between IL-6 and TGF-β and between Th17 and Treg in a Chronic Stress-Induced Mouse Model of Depression. Int. J. Mol. Sci. 2022, 23, 13856.
  189. Mihailova, S.; Ivanova-Genova, E.; Lukanov, T.; Stoyanova, V.; Milanova, V.; Naumova, E. A Study of TNF-α, TGF-β, IL-10, IL-6, and IFN-γ Gene Polymorphisms in Patients with Depression. J. Neuroimmunol. 2016, 293, 123–128.
  190. Cattaneo, A.; Cattane, N.; Malpighi, C.; Czamara, D.; Suarez, A.; Mariani, N.; Kajantie, E.; Luoni, A.; Eriksson, J.G.; Lahti, J.; et al. FoxO1, A2M, and TGF-Β1: Three Novel Genes Predicting Depression in Gene X Environment Interactions Are Identified Using Cross-Species and Cross-Tissues Transcriptomic and MiRNomic Analyses. Mol. Psychiatry 2018, 23, 2192–2208.
  191. Wyller, V.B.; Nguyen, C.B.; Ludviksen, J.A.; Mollnes, T.E. Transforming Growth Factor Beta (TGF-β) in Adolescent Chronic Fatigue Syndrome. J. Transl. Med. 2017, 15, 245.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , ,
View Times: 392
Revisions: 2 times (View History)
Update Date: 31 Jan 2023
1000/1000