Next Article in Journal
Impact of Antiglaucoma Drug Number and Class on Corneal Epithelial Thickness Measured by OCT
Previous Article in Journal
Acute Compartment Syndrome and Intra-Abdominal Hypertension, Decompression, Current Pharmacotherapy, and Stable Gastric Pentadecapeptide BPC 157 Solution
Previous Article in Special Issue
An Overview of the Systematic Reviews About the Efficacy of Fluvoxamine on Depression
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Unveiling the Anti-Inflammatory Effects of Antidepressants: A Systematic Review of Human Studies over the Last Decade

by
Layla Bleibel
1,*,
Paulina Sokołowska
1,
Gabriela Henrykowska
2,
Jacek Owczarek
3 and
Anna Wiktorowska-Owczarek
1,*
1
Department of Pharmacology and Toxicology, Medical University of Lodz, Żeligowskiego 7/9, 90-752 Lodz, Poland
2
Department of Epidemiology and Public Health, Medical University of Lodz, Żeligowskiego 7/9, 90-752 Lodz, Poland
3
Department of Hospital Pharmacy, Medical University of Lodz, Muszyńskiego 1, 90-151 Lodz, Poland
*
Authors to whom correspondence should be addressed.
Pharmaceuticals 2025, 18(6), 867; https://doi.org/10.3390/ph18060867
Submission received: 25 March 2025 / Revised: 5 June 2025 / Accepted: 6 June 2025 / Published: 10 June 2025
(This article belongs to the Special Issue Pharmacology of Antidepressants: Recent Advances)

Abstract

:
Background/Objectives: Depression ranks among the most prevalent mental health conditions globally, marked by a variety of symptoms that frequently cause significant emotional distress and impairment in individuals, alongside a high recurrence rate. The predominant approach to treating depression revolves around monoamine theory, utilizing SSRIs and SNRIs, with Esketamine emerging as a supplementary option in recent times. Nevertheless, there is a growing focus on exploring the relationship between inflammation and depression, revealing a strong correlation between the two. This insight prompts consideration of the anti-inflammatory properties of current antidepressants in their therapeutic application. Methods: A systematic literature search was conducted using the PubMed database to identify randomized controlled trials (RCTs) and clinical trials (CTs) that assessed the in vivo anti-inflammatory effects of SSRIs (fluoxetine, escitalopram, sertraline, and paroxetine), the SNRI venlafaxine, and esketamine/ketamine in human subjects undergoing treatment for depression. The included studies were evaluated based on changes in levels of pro-inflammatory and anti-inflammatory markers in response to the antidepressant treatments. Results: SSRIs, SNRIs, esketamine, and ketamine (a racemic mixture of S- and R-ketamine not formally approved for the treatment of depression) exhibit anti-inflammatory effects through diverse mechanisms, such as reducing pro-inflammatory cytokines or enhancing anti-inflammatory cytokines in serum or within specific brain regions like the hippocampus and prefrontal cortex. These actions are mediated through various inflammatory pathways, including nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), the brain Nod-like receptor pyrin-containing 3 (NLRP3) inflammasome, the glutamatergic system, the gut–brain axis, the hypothalamic–pituitary axis, impaired neuroplasticity, and the kynurenine pathway. Conclusions: In summary, SSRIs, SNRIs, esketamine, and ketamine exert an anti-inflammatory role alongside their antidepressant effects via these intricate mechanisms.

1. Introduction

Major depressive disorder (MDD), commonly referred to as depression, stands as the most prevalent and highly debilitating mental health condition worldwide, affecting 3.8% of the global population, with a notable impact on 5% of adults, as highlighted by the World Health Organization [1]. This disorder takes a significant socioeconomic toll, with only a fraction—approximately 30% to 35%—of affected adults achieving remission through current therapeutic approaches [2,3,4]. Moreover, the recurrence rate following the initial major depressive episode is as high as 60% [5]. Projections estimate that by 2030, depression will be the second most serious global health condition [6]. Furthermore, after the coronavirus disease (COVID) in 2020, cases of major depression increased by more than a quarter (28%) worldwide [7].
MDD pathogenesis is complex and most likely multi-factorial with various mechanisms interacting and affecting one another. Several hypotheses have been proposed to explain its pathogenesis, including the monoamine hypothesis, the cytokine hypothesis, the neuroplasticity hypothesis, and the hypothalamic–pituitary–adrenal (HPA) axis hypothesis [8,9,10,11].
The primary treatment of depression is derived from the monoamine hypothesis. Following the incidental discovery that two tuberculosis drugs alleviated the symptoms of depression, the monoamine theory of depression was born, around 50 years ago [12,13]. This theory hypothesized that the deficiency of noradrenergic and or/serotonergic neurotransmitters is the basis of the cause of depression, and replenishing these neurotransmitters can act as a treatment for depression [14,15].
In the late 1960s, the mechanisms of serotonin degradation, synthesis, and reuptake were being characterized. Then, in the early 1970s, based on trying to increase serotonin neurotransmission in the synaptic cleft, the first serotonin reuptake inhibitor (SSRI) fluoxetine was introduced [16,17]. Clinical trials lasted more than 7 years, although it was approved by the US Food and Drug Administration (FDA) in 1987 [18]. The first serotonin–norepinephrine reuptake inhibitor (SNRI), venlafaxine, was introduced in 1993 in the United States market [19].
However, recent studies have suggested that the monoamine hypothesis of depression should be revised, as the pathogenesis of MDD is still unclear and it does not provide a clear explanation of the action of antidepressants [15]. Moreover, they are effective in less than 50% of patients [20], and require chronic treatment to induce an effect [21,22]. The revised monoamine hypothesis suggests that monoamines play a modulatory role on other neurobiological systems that have a more primary role in depression or that they must be present in the context of stressors [23,24].
Clinical research has uncovered evidence indicating dysfunction in the glutamatergic system among patients with MDD. This includes findings such as increased concentrations of glutamate in plasma [25,26,27] and increased concentrations of glutamine in cerebrospinal fluid (CSF) [28]. Ketamine, originally developed as a dissociative anesthetic in the 1960s and FDA-approved in 1970 [29], is a noncompetitive N-methyl-D-aspartate (NMDA) receptor antagonist [30,31,32]. Additionally, it exhibits weak binding to sigma [33], muscarinic [34], and κ and δ opioid receptors [35], as well as to dopamine, norepinephrine, and serotonin transporters [36]. Ketamine blocks NMDA receptors, leading to calcium influx and the subsequent release of brain-derived neurotrophic factor (BDNF). This release activates pathways that promote synaptogenesis and neurogenesis, ultimately reducing depressive symptoms [37].
Overall, clinical data suggests that the glutamatergic system plays a role in the pathophysiology of MDD, involving disruptions in glutamatergic substrate concentrations and alterations in NMDA receptors [38,39,40,41]. Recent clinical breakthroughs have led to the FDA approval of esketamine, the S-enantiomer of ketamine, as a nasal spray for adjunctive treatment in patients with treatment-resistant depression, signifying the growing recognition of its therapeutic potential. However, it is important to emphasize that this approval applies exclusively to esketamine, not to racemic ketamine. In our current study, we examine the effects of both compounds [42] because ketamine has been widely studied for its anti-inflammatory and antidepressant properties.
Given the dominating monoamine theory of depression and the widespread use of SSRIs and SNRIs as primary treatments, along with esketamine’s role in treating treatment-resistant depression, it is crucial to explore their potential anti-inflammatory effects. This is especially important as emerging theories, such as the link between inflammation and depression, are gaining attention [43]. Understanding how antidepressants influence inflammatory pathways, particularly their effects on pro- and anti-inflammatory cytokines, may offer valuable insights into their therapeutic mechanisms. The aim of this systematic review is to evaluate the anti-inflammatory effects of antidepressants from the SSRI, SNRI, and esketamine (including ketamine) classes, specifically focusing on their impact on cytokine levels as a marker of the inflammatory response. This review will analyze human studies that have quantitatively assessed changes in cytokine levels—whether an increase, decrease, or no effect—in response to antidepressant administration.

2. Relationship Between Inflammation and Depression

Before exploring the link between antidepressants and inflammation, it is essential to first examine the connection between inflammation and the pathogenesis of depression. Infection, chronic stress, and stress-inducing events that cause a systemic inflammatory response are widely recognized as significant contributors to the pathogenesis of depression [44,45]. The inflammatory process and immune system activation in depression have been observed in both the peripheral and central nervous systems (CNS). More specifically, communication between peripheral inflammation and the CNS was identified to be mediated mainly by the nuclear factor (NF-κB) at the blood–brain barrier [46,47]. Interleukine1 beta (IL-1β) [48], Tumor Necrosis Factor Alpha (TNF-α) [49], and Interleukine1 (IL-1) are the primary systemic inflammatory cytokines inducing the body’s inflammatory response and serve as important biomarkers of inflammation [11]. TNF-α release into the bloodstream initiates a cascade of immune system activation and release of additional pro-inflammatory cytokines such as IL-8, IL-1β, and IL-6 [50]. This systemic inflammatory response leads to a disruption of the blood–brain barrier (BBB), a highly selective and semi-permeable membrane that protects the brain and CNS from pathogens. However, systemic inflammation causes a disruption in the integrity and permeability of the membrane which allows inflammatory mediators to reach the brain and trigger a neuroinflammatory reaction [51].
The association between inflammation and depression was initially discovered when multiple studies found that administering IL-2 and IFN-gamma to patients triggered depressive symptoms such as anhedonia and fatigue [52,53,54,55,56,57,58]. Elevated levels of pro-inflammatory cytokines were found in patients with MDD, even though they had no underlying somatic disease [59] (Figure 1). Furthermore, growing evidence suggests that individuals with depression also exhibit elevated levels of circulating cytokines like IL-6 [49,60,61], IL-1β [48], TNF-α [49], IFN-α, prostaglandin E2 (PGE2), and chemokine CCL2 [49,62,63,64,65], as well as acute-phase proteins such as C-reactive protein (CRP) [66]. This relationship has also been observed in patients with systemic diseases that have an inflammatory pathophysiology such as diabetes mellitus type 1 [67], inflammatory bowel disease, and rheumatoid arthritis [68]. Moreover, reduced levels of the anti-inflammatory cytokines transforming growth factor (TGF-β) and IL-10 were observed in the brain [69]. These factors all indicate the presence of chronic low-grade inflammation in MDD [70]. Patients with higher levels of inflammatory factors have also been correlated with a decreased response to antidepressant treatment [71], precisely SSRIs, and a better response to an SNRI or an add-on regimen with bupropion or anti-inflammatory agents [72,73] (Figure 1).

2.1. Mechanisms of Inflammation

Peripheral inflammation can contribute to depressive symptoms by reaching the CNS through various mechanisms, including the gut-microbiota–brain axis, the HPA axis, the glutamatergic system, and the kynurenine (KYN) pathway, as well as by impairing neuroplasticity [74,75,76,77].
Chronic stress leads to the activation of macrophages and monocytes, which in turn trigger the release of pro-inflammatory cytokines such as IL-1β, IL-1, and IL-6 [78,79]. These peripheral pro-inflammatory cytokines can reach the CNS and stimulate glial cells and astrocytes. Through a feedback mechanism, these CNS immune cells then produce cytokines [80], leading to neuroinflammation, which is likely to play a significant role in the development of depressive symptoms [61,81,82]. This process is thought to be mediated by peripheral macrophages, as studies inhibiting these cells have shown reduced levels of pro-inflammatory cytokines [83,84]. This was known as the macrophage theory of depression, and it was first proposed by Smith et al. in 1991 [85].

2.2. Neuroinflammation

2.2.1. Microglia

Microglia are the main immune cells in the CNS, and it was found that in models of neurodegeneration, inflammation, and aging, microglia favor inflammation marker activation [86,87,88]. Then, pro-inflammatory mediators were found to be increased in various causes of depression, such as inflammation, gut dysbiosis, or stress, all of which have been associated with persistent activation of microglia [89,90,91,92,93]. Furthermore, in a neuroinflammatory state, microglia exhibit reduced expression of neuroprotective genes like nerve growth factor (NGF), neurotrophins (NTs) 4/5, glial cell-derived neurotrophic factor (GDNF), and brain-derived neurotrophic factor (BDNF). Under non-inflammatory conditions, these genes offer vital trophic support to neurons, and their reduced expression can impair neurogenesis and synaptic plasticity [94], potentially leading to depressive-like symptoms [95]. It has been suggested that alterations in neurotrophins, including the decreased expression of BDNF observed in the brains of patients with MDD, play a key role in the impaired neuroplasticity. Additionally, pro-inflammatory cytokines further impact BDNF levels in the brain [96,97,98,99]. Studies found that the anti-inflammatory agent minocycline has a neuroprotective effect by decreasing the pro-inflammatory proliferation of microglia [100,101,102].

2.2.2. Astrocytes

Similar to microglia, astrocytes contribute to proper neuronal development by releasing growth factors such as BDNF, vascular endothelial growth factor (VEGF), nerve growth factor (NGF), ciliary neurotrophic factor (CNTF), and fibroblast growth factor 2 (FGF2), also known as basic FGF (bFGF) [103]. Moreover, their function may be compromised during an inflammatory state, affecting their ability to maintain CNS homeostasis and consequently impacting neuronal survival [104]. Elevated plasma levels of glial fibrillary acidic protein (GFAP) and S100β, markers of astrocytic activation, have been observed in patients with treatment-resistant depression compared to healthy individuals [105].

2.2.3. Lymphocyte Infiltration

Recent studies have demonstrated that lymphocytic infiltration can occur in the CNS under specific conditions, challenging the long-standing concept of the CNS as an immune-privileged site [106]. A study conducted by Schlaaf et al., which involved mapping T and B lymphocytes across whole-brain sections, identified distinct patterns of neuroinflammation, characterized by increased densities of T and/or B cells, within the white matter, cortical areas, and limbic system in a significant subset of individuals with schizophrenia and mood disorders. These findings lend support to the hypothesis that lymphocyte infiltration occurs at a greater percentage in patients with mood disorders compared to healthy controls. The observed increase in lymphocyte density suggests a compromise of the blood–brain barrier, as under normal physiological conditions, lymphocytes rarely cross this barrier [107].

2.2.4. Neuroplasticity

Neuroplasticity refers to the ability of the CNS to adapt and modify itself in response to various stimuli, by restructuring and/or rewiring its neural connections [108,109,110,111]. Changes in gray matter volume, measurable through MRI in clinical studies, serve as an important indicator of neuroplasticity [111]. Among brain regions, the hippocampus has been the most extensively studied in the context of depression, with evidence suggesting that stress and other adverse stimuli can impair hippocampal plasticity [112,113].
Neuroimaging studies demonstrate reductions in gray matter volume in MDD, particularly in the hippocampus [114,115] and anterior cingulate cortex [116]. Postmortem analyses found further neuronal and glial loss, particularly in individuals with chronic forms of the illness [117,118]. These observed decreases in the volumes of the prefrontal cortex and hippocampus are thought to result from neuronal and glial disruption and atrophy associated with depression [119,120]. These observations support the neuroplasticity hypothesis of MDD, which proposes that impaired neuroplasticity plays a central role in the pathogenesis of depression [121].
Antidepressants have been shown in experimental studies to enhance neuroplasticity, stimulate hippocampal neurogenesis, and mitigate the deleterious effects of stress on the hippocampus [122,123,124,125]. Supporting these findings in humans, postmortem studies have shown that patients with depression receiving antidepressant treatment exhibit greater total dentate granule cell numbers and increased dentate gyrus volume compared to nonmedicated individuals [126]. While these effects appear to be important in certain models of depressive-like behavior, evidence also indicates that antidepressant efficacy may also involve neurogenesis-independent mechanisms [127].

2.2.5. Oxidative Stress

Reactive oxygen species (ROS) play an important role in not only the pathogenesis of neurological disease but also in normal brain function [128]. Oxidative stress (OS) is characterized by increased production of ROS (free radicals) in the cells and tissues and simultaneously the exhaustion of antioxidative defenses, which causes an imbalance and inability to neutralize ROS. This leads to damage to DNA, lipids and proteins [129,130]. The brain is particularly susceptible to ROS-induced injury due to its high oxygen metabolism and relatively weak antioxidant defenses. Consequently, ROS produced by microglia and astrocytes in brain tissue can trigger neuroinflammation, cell death, and subsequent neurodegeneration and memory loss [131]. Furthermore, ROS-mediated lipid peroxidation in the brain leads to the generation of toxic compounds that induce neuronal cell death [132], representing a significant contributing factor to the pathogenesis of MDD [133].
Therefore, an increase in ROS production and a decreased antioxidant response lead to neurodegeneration, inflammation, tissue damage, and cell death [134]. These processes have been linked to the pathogenesis and progression of MDD [135,136] and it is known as the ‘oxidative stress hypothesis of depressive disorders’ [137,138,139]. Increased ROS levels were also found to modulate feedback in the HPA axis and to effect serotonergic and GABA transmission in experimental rodent models of anxiety and depression [140]. OS is then increased by HPA axis activation [141].

2.2.6. Hypothalamic–Adrenal Axis

Approximately 40% to 60% of patients with MDD experience disturbances in the HPA axis, such as hypercortisolism, mirroring the effects of chronic stress [142]. This can be triggered by HPA axis activation due to increased cytokine production, which can also lead to glucocorticoid receptor resistance [143]. This stress and HPA axis dysregulation result in functional changes not only in the hippocampus [144] but also in the amygdala and prefrontal cortex [145,146]. Stress increases the release of adrenocorticotropic hormone (ACTH), corticotropin-releasing factor (CRF), and corticosterone (CORT) in rats [147]. Increased levels of CORT have also been found to increase NLRP-3 inflammasome levels in the hippocampus, stimulating the pro-inflammatory response [148,149]. Furthermore, increased levels of glucocorticoids for a long period of time also lead to neuronal death, synapse loss, and changes in neuronal dendrites [150]. Additionally, suppression of the HPA axis was seen in a study of patients with depression being treated with antidepressants, decreasing the stress response [151,152].

2.2.7. The Kynurenine Pathway

The kynurenine pathway is intricately regulated by the immune system and has a significant connection to the central nervous system (CNS). Within this pathway, two mechanisms are believed to contribute to inflammation. The first is the tryptophan starvation hypothesis, which suggests that activation of the kynurenine pathway decreases levels of tryptophan as it is converted to kynurenine, which is mediated by indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO) [153], reducing its availability for serotonin synthesis. The second mechanism involves a shift within the kynurenine metabolism pathway toward producing neurotoxic metabolites, particularly quinolinic acid [154]. Quinolinic acid, generated primarily by activated monocytic cells, induces excitotoxicity, stimulates the production of reactive oxygen species (ROS), increases lipid peroxidation in the CNS, and disrupts glutamatergic neurotransmission. This disruption occurs as kynurenic acid and NMDA receptors block glutamate receptors, therefore contributing to excitotoxicity, neuroinflammation, and oxidative stress [155,156,157,158,159]. This is supported by a study on patients with major depression and suicidal behavior, which revealed decreased CSF levels of kynurenine and increased levels of the neurotoxin quinolinic acid compared to healthy individuals [160]. These findings underscore the kynurenine pathway’s role in neurodegeneration, behavior, and overall neural function [159].

2.2.8. Gut–Brain Axis

Recent studies have highlighted the critical relationship between the gut microbiota and the brain, commonly referred to as the gut–brain axis [161,162]. This relationship is bidirectional and mediated through multiple pathways, including the hypothalamic–adrenal axis [163], the autonomic and enteric nervous systems, the immune system [164], the endocrine system, and the gut microbiota and its metabolites [165,166,167,168,169,170]. The latest theories also suggest that bacterial extracellular vesicles may travel to the CNS, exerting their effects there [171].
The gut microbiota plays a vital role not only in maintaining overall host health but also in supporting brain health and development. An imbalance between beneficial and pathogenic bacteria in the gut, known as dysbiosis, can disrupt CNS function [172,173,174]. Dysbiosis has been shown to increase inflammatory cytokines, which alter the permeability of both the blood–brain barrier and the gut barrier, leading to neuroinflammation [175]. Stress further disrupts gut microbiota balance, influencing immune mechanisms and promoting microglial activation and cytokine secretion [176].
This connection is particularly evident in patients with chronic conditions such as inflammatory bowel disease (IBD), who frequently exhibit concurrent MDD [177]. These findings highlight the role of chronic gut inflammation in the development of MDD symptoms [178]. Additionally, a study demonstrated that daily administration of low-dose tricyclic antidepressants produced an 85% moderate response rate in patients with IBD, highlighting the therapeutic potential of targeting the gut–brain axis [179].
Various studies also indicate that probiotics may help alleviate the symptoms in patients with psychiatric disorders, providing further evidence of the gut microbiota’s impact on mental health [180,181,182,183,184,185,186,187,188]. These findings highlight the critical role of the gut–brain axis in the development of inflammation and consequently MDD symptoms, further adding to the mechanisms linking inflammation and depression.

3. Results

3.1. Anti-Inflammatory Effect of Antidepressants

3.1.1. IL-1β

IL-1β, a cytokine belonging to the IL-1 family, plays a crucial role in the host’s defense against pathogens, particularly in injury and infection, and exhibits potent pro-inflammatory activity [189]. Increased levels of IL-1β have been linked to an increase in Hamilton depression rating scale (HAMD) scores [190]. In our review, we noticed that IL-1β decreased with the administration of the SSRIs escitalopram [191] (one study) and fluoxetine [192,193,194] (three studies) in patients with depression, as well as with ketamine [195] (one study in patients with treatment-resistant depression). However, one study on patients with MDD given fluoxetine showed no significant effect on IL-1β serum levels [196] (Table 1 and Figure 2).

3.1.2. IL-1ra

IL-1ra, also a member of the IL-1 cytokine family, functions as an IL-1 receptor antagonist. By binding to the same receptors as IL-1β and IL-1α, it effectively blocks these cytokines, thereby modulating their pro-inflammatory effects in the human body [197]. Furthermore, studies have shown that it is an effective anti-inflammatory against various diseases such as colitis and arthritis. In our review, we noticed that IL-1ra decreased with the administration of the SSRIs escitalopram [191] and fluoxetine [193] in patients with depression. However, one study on patients with MDD given escitalopram [198] showed no significant effect on IL-1ra serum levels in patients with depression (Table 1 and Figure 2).

3.1.3. IL-2

IL-2 is a cytokine with extensive pro-inflammatory roles, including promoting the differentiation of CD8+ T cells into cytotoxic T lymphocytes [199,200,201] and guiding the maturation of CD4+ naive T cells into regulatory T cells within the thymus [202]. In our review, we noticed that IL-2 decreased with the administration of the SSRIs escitalopram [191,203] (one study in patients with moderate depression, another study in patients with MDD), fluoxetine (one study, patients with MDD) [203], sertraline (one study, patients with unipolar depression) [204], as well as with ketamine (one study, patients with treatment-resistant depression) [195]. However, one study on patients with MDD given fluoxetine showed no significant effect on IL-2 serum levels [205] (Table 1 and Figure 2).

3.1.4. IL-4

IL-4 is a cytokine with immunoregulatory functions, playing a role in modulating inflammation, supporting hematopoiesis, and regulating antibody production [206]. It exerts its anti-inflammatory effect by inhibiting the production of IL-1β and TNF-α [207] and also inducing the production of IL-1ra [208]. In our review, we noticed that IL-4 decreased with the administration of the SSRIs escitalopram (one study, moderate depression) [191] and sertraline (one study, unipolar depression) [204], as well as with ketamine (one study, patients with treatment-resistant depression) [195]. Meanwhile, it increased with the SSRI fluoxetine (two studies, first on depressed patients [193], only at week 8, second on first-episode adolescent patients with moderate to severe major depressive disorder [194]) (Table 1 and Figure 2).

3.1.5. IL-5

IL-5 is a pro-inflammatory cytokine that stimulates the maturation, proliferation, and migration of eosinophils [209,210]. In our review, we noticed that IL-5 decreased with the administration of the SSRIs escitalopram (one study, moderate depression) [191] and with ketamine (one study, treatment-resistant depression) [195]. However, one study [193] classifies IL-5 as an anti-inflammatory cytokine rather than a pro-inflammatory one. Its findings showed an increase in IL-5 levels after administration of the SSRI fluoxetine in depressed patients [193] (Table 1 and Figure 2).

3.1.6. IL-6

IL-6 plays a key role in host defense against pathogens, injury, and infection. Its pro-inflammatory mechanism lies in stimulating immune reactions [211,212], acute-phase responses [213], and hematopoiesis [214]. In our review, we noticed that IL-6 decreased with the administration of the SSRIs escitalopram (three studies, moderate depression [191] and MDD [203,215]), sertraline (three studies, first hemodialysis and depression [216], second unipolar depression [204], and last congestive heart failure and depression [217]), and fluoxetine (two studies, MDD [203] and depression [193]), with ketamine (one study, treatment-resistant depression [195]), with esketamine (two studies, labor and postpartum depression [218], and elective non-cardiac thoracic surgery with high-dose esketamine [219]), while it increased with paroxetine (one study, MDD) [220] and ketamine (one study, MDD) [221].
However, one study on patients with MDD given fluoxetine showed no significant effect on IL-6 serum levels [196]. Another study with escitalopram showed no significant effect on IL-6 serum levels in patients with depression [198]. Two studies with sertraline also showed no effect, the first on patients with MDD and CKD [222] and the second on patients with CHD and comorbid depression [223] (Table 1 and Figure 2).

3.1.7. IL-7

IL-7 has a vital role in various aspects of the immune system, and in inflammation, it acts by promoting pro-inflammatory cells and cytokines [224]. In our review, we noticed that IL-7 decreased with the administration of the SSRI escitalopram (one study, moderate depression [191]) and ketamine (one study, treatment-resistant depression [195]) (Table 1 and Figure 2).

3.1.8. IL-8

IL-8 is a pro-inflammatory chemokine distinguished from other cytokines by its high affinity for signaling and attracting neutrophils [225]. In our review, we noticed that IL-8 decreased with the administration of the SSRI escitalopram (one study, moderate depression [191]) and with ketamine (one study, depression [226]). However, one study on patients with MDD given escitalopram showed no significant effect on IL-8 serum levels in patients with depression [198] (Table 1 and Figure 2).

3.1.9. IL-9

IL-9 is primarily involved in promoting mast cell activity and regulating IgE production [227]; these are linked to the pathogenesis of asthma and protecting the body against parasitic infections [228]. In our review, we noticed that IL-9 decreased with the administration of the SSRI escitalopram (one study, moderate depression) [191] (Table 1 and Figure 2).

3.1.10. IL-10

IL-10 is a cytokine essential for its anti-inflammatory functions, playing a key role in regulating inflammation and preventing autoimmune activation in the body. It works by suppressing the activity of T cells and macrophages [229,230]. In our review, we noticed that IL-10 decreased with the administration of the SSRI escitalopram (one study, moderate depression) [191] and ketamine (one study, treatment-resistant depression) [195], and it increased with the SSRI fluoxetine (one study, Crohn’s disease) [231], paroxetine (one study, MDD) [220], and esketamine (one study, labor and postpartum depression) [218]. However, in another study with sertraline, IL-10 decreased (unipolar depression) [204], and another study on patients with MDD given escitalopram showed no significant effect on IL-10 serum levels [198] and a non-statistically significant increase with sertraline (one study, hemodialysis patients with depression) [216] (Table 1 and Figure 2).

3.1.11. IL-12

IL-12 uniquely regulates inflammation by influencing both the innate and adaptive immune systems. It stimulates T lymphocytes to produce IFN-γ [232] and also supports the cytotoxic activity of T cells and NK cells by promoting them to release perforins and granzymes [233,234,235,236]. In our review, we noticed that IL-12 increased with the administration of the SSRIs escitalopram (one study, moderate depression) [191] and fluoxetine (one study, depressed patients, week 8) [193], while it decreased with fluoxetine (one study, depressed patients, week 4) [193] (Table 1 and Figure 2).

3.1.12. IL-13

IL-13 is primarily an anti-inflammatory cytokine that influences monocytes and B cells, reduces the production of pro-inflammatory cytokines, and induces class switching to IgE [237,238]. These actions contribute to its role in mediating allergic asthma. In our review, we noticed that IL-13 decreased with the administration of the SSRI escitalopram (one study, moderate depression [191]) (Table 1 and Figure 2).

3.1.13. IL-15

IL-15 plays a crucial pro-inflammatory and immunomodulatory role in defending against various pathogens—including viruses, bacteria, and parasites—by activating and stimulating lymphocytes [239]. In our review, we noticed that IL-15 increased with the administration of the SSRI escitalopram (one study, moderate depression [191]), while it decreased with fluoxetine (one study, depressed patients, week 4) [193] (Table 1 and Figure 2).

3.1.14. IL-17

IL-17 is a pro-inflammatory cytokine that promotes the expression of IL-6 and granulocyte colony-stimulating factor (G-CSF), which in turn enhances neutrophil recruitment to the site of infection and stimulates granulopoiesis [240]. In our review, we noticed that IL-17 decreased with the administration of the SSRIs escitalopram (one study, moderate depression [191]) and sertraline (one study, patients with unipolar depression [204]) (Table 1 and Figure 2).

3.1.15. IFN-γ

IFN-γ, produced by T lymphocytes and NK cells, exerts powerful immunomodulatory effects by regulating cell proliferation and apoptosis, as well as activating the innate immune system and macrophages [241,242]. In our review, we noticed that IFN-γ decreased with the administration of the SSRIs escitalopram (one study, moderate depression) [191], sertraline (one study, patients with unipolar depression) [204], and fluoxetine (one study, depressed patients, week 4) [193], as well as ketamine (one study, patients with treatment-resistant depression) [195] (Table 1 and Figure 2).

3.1.16. TNF-α

TNF-α is a cytokine with diverse functions, primarily responsible for promoting inflammation and contributing to the pathogenesis of autoimmune diseases. Additionally, its signaling pathways can lead to cellular apoptosis and necrosis [243,244,245]. In our review, we noticed that TNF-α decreased with the administration of the SSRI escitalopram (two studies, moderate depression [191] and MDD [203]) and ketamine (two different studies on treatment-resistant depression [195,246]). A third study on patients with treatment-resistant depression showed an increase in TNF-α serum levels with ketamine [247]. However, one study on patients with MDD given escitalopram showed no significant effect on TNF-α serum levels in patients with depression [198], and two studies with sertraline also showed no effect (first congestive heart disease and comorbid depression [223], and second on unipolar depression [204]). With the SSRI paroxetine, TNF-a levels were increased in patients with MDD [220] (Table 1 and Figure 2).

3.1.17. CRP

C-reactive protein (CRP) is an acute-phase protein released in response to inflammation and tissue damage [248]. As a result, CRP levels are commonly used to monitor infection, inflammation, and autoimmune diseases [249,250]. In our review, we noticed that CRP decreased with the administration of the SSRI sertraline (one study, congestive heart failure and depression [217]), the SNRI venlafaxine (one study, patients with MDD [220]), ketamine (one study, patients with post-operative cognitive dysfunction after cardiac surgery [251]), and esketamine (one study, labor and postpartum depression [218]). CRP levels were increased in patients with MDD with the SSRI paroxetine [220]. Two studies with sertraline also showed no effect on CRP levels (first on MDD and chronic kidney disease [222], and second on congestive heart disease and comorbid depression [223]) (Table 1 and Figure 2).
Table 1. The effect (decrease (↓), increase (↑), or no statistically significant effect (↔)) of antidepressants (SSRIs, SNRIs, ketamine, and esketamine) on inflammatory and non-inflammatory factors; all studies were conducted in serum. TNF-α: Tumor Necrosis Factor Alpha; NLRP3: Nod-like receptor pyrin-containing 3; IFN-γ: interferon gamma; hsCRP: high-sensitivity C-reactive protein; MDD: major depressive disorder; TRD: treatment-resistant depression; CKD: chronic kidney disease; LPS: lipopolysaccharide; PHA: phytohemagglutinin; CHD: coronary heart disease.
Table 1. The effect (decrease (↓), increase (↑), or no statistically significant effect (↔)) of antidepressants (SSRIs, SNRIs, ketamine, and esketamine) on inflammatory and non-inflammatory factors; all studies were conducted in serum. TNF-α: Tumor Necrosis Factor Alpha; NLRP3: Nod-like receptor pyrin-containing 3; IFN-γ: interferon gamma; hsCRP: high-sensitivity C-reactive protein; MDD: major depressive disorder; TRD: treatment-resistant depression; CKD: chronic kidney disease; LPS: lipopolysaccharide; PHA: phytohemagglutinin; CHD: coronary heart disease.
NameClassEffectCytokinesModelCitation
EscitalopramSSRIIL-1β, IL-2, IL-5, IL-6, IL-7, IL-8, IL-9, IL-17, TNF-α, IFN-γModerate depression,
patients were their own control, serum, Luminex xMAP multiplexing technology, 26 weeks of antidepressant treatment, 90 participants (71% women) with a mean age of 38 years.
[191]
IL-1ra, IL-4, IL-10, IL-13[191]
IL-12, IL-15[191]
IL-10, IL-1ra, IL-6, IL-8, TNF-αDepression, randomized double-blinded trial, whole blood stimulated with LPS or PHA (in vitro), Luminex 100 platform, 4 weeks of antidepressant treatment, 44 participants (28 women), with a mean age of 32.7 years.[198]
IL-2, IL-6 and TNF-aMDD, patients were their own control, cytokines were analyzed in
serum by ELISA test, 6 weeks of antidepressant treatment, 65 participants (35 women), with a mean age of 36 years.
[203]
IL-6MDD, open-label part-randomized multicenter pharmacogenetic study with two active pharmacological
treatment arms, leukocyte mRNA levels were measured, 8 weeks of
antidepressant treatment, 38 participants with a mean age of 38 years.
[215]
FluoxetineIL-1β, IL-6MDD, patients were their own control, cytokines were analyzed in
serum by ELISA test, 8 weeks of study, 14 participants (10 women), with a mean age of 37 years.
[196]
IL-2, IL-6 and TNF-aMDD, patients were their own control, cytokines were analyzed in
serum by ELISA test, 6 weeks of study, 65 participants (36 women), with a mean age of 36 years.
[203]
IL-10Crohn’s disease, presence of a separate control group that received a placebo, cytokines were analyzed by flow cytometry, 6 months of antidepressant treatment, 26 participants (12 women), with a mean age of 37 years.[231]
TNF-aDepression, patients were their own control, cytokines were analyzed in serum by ELISA test, 12 weeks of antidepressant treatment, 30 participants (20 women), with a mean age of 36 years.[252]
IL-1βDepression, the effect of fluoxetine was compared to the untreated group, cytokines were analyzed in serum by ELISA test, 12 weeks of antidepressant treatment, 32 participants (18 women), with a mean age of 34 years.[192]
IL-2, CRPDepression[205]
IL-1raDepression, cytokine measurement in the 4th week of therapy,
presence of a separate control group, cytokines were analyzed in
serum by multiplex bead-based immunoassays, 8 weeks of antidepressant treatment, 22 participants (18 women), with a mean age of 17 years.
[193]
IFN-γ, IL-1β, TNF-α, IL-6, IL-12, IL-15[193]
IL-4 Depression, cytokine measurement in the 8th week of therapy.[193]
IL-12 and IL-5[193]
IL-10 and IL-13Depression[193]
CRPCOVID-19; during a mid-hospital stay, a double-blind randomized, placebo-controlled
clinical trial; 4 weeks of antidepressant treatment, 72 participants (35 women), with a mean age of 52 years.
[253]
NLRP3, IL-1β, and IL-18First episode, moderate to severe MDD, patients were their own control, cytokines were analyzed in serum by ELISA test, 12 weeks of antidepressant treatment, 48 participants (30 women), with a mean age of 17 years.[194]
IL-4 [194]
SertralinehsCRP, IL-6MDD and CKD, randomized, double-blind placebo-controlled trial, cytokines were analyzed in serum by ELISA test, 12 weeks of antidepressant treatment, 201 participants (61 women), with a mean age of 58 years.[222]
IL-6Hemodialysis and depression, randomized double-blind, placebo-controlled clinical trial, cytokines were analyzed in serum by ELISA test, 12 weeks of antidepressant treatment, 43 participants (16 women), with a mean age of 63 years.[216]
TNF-α [216]
IL-10[216]
IL-4, IL-10Unipolar depression, double-blind, placebo-controlled trial, blood cytokines were measured by flow cytometry, 6 weeks of antidepressant treatment, 120 participants (82 women), with a mean age of 42 years.[204]
IL-2, IL-6, IL-17a, IFN-γ[204]
TNF-α [204]
hsCRP, IL-6, and TNF-α CHD and MDD, randomized, double-blind,
placebo-controlled trial, cytokines were analyzed in serum by ELISA test, 10 weeks of antidepressant treatment, 122 participants (41 women), with a mean age of 59 years.
[223]
CRP and IL-6 CHD and MDD, randomized double-blind, placebo-controlled clinical trial, cytokines were analyzed in serum by ELISA test, 20 weeks of antidepressant treatment, 95 participants (48 women), with a mean age of 57 years.[217]
ParoxetineIL-10MDD, two randomized placebo-controlled clinical studies, cytokines were analyzed in serum by ELISA test, 10 weeks of antidepressant treatment, 106 participants (72 women), with a mean age of 46 years.[220]
TNF-α, IL-6 and CRP[220]
VenlafaxineSNRICRPMDD, two randomized placebo-controlled clinical studies, cytokines were analyzed in serum by ELISA test, 10 weeks of antidepressant treatment, 104 participants (64 women), with a mean age of 45 years.[220]
Ketamine CRPPost-operative cognitive dysfunction, patients randomly received placebo or an i.v. bolus of ketamine (0.5 mg/kg) during anesthetic induction. Anesthesia was maintained with isoflurane and fentanyl. A nonsurgical group was also
included as control, serum C-reactive protein (CRP) concentrations were determined before surgery and on the first post-operative day, 96 participants with a mean age of 66 years.
[251]
IL-8/IL-10 ratioMDD, presence of a separate control group that received a placebo, plasma concentrations of cytokines were analyzed by ELISA test 24 h after ketamine infusion (0.5 mg/kg), 25 participants (7 women) with a mean age of 46 years.[254]
IL-4, and IL-10TRD, cytokines were analyzed in plasma by multiplex bead-based immunoassays on days 13 and 26, TRD patients received intravenous ketamine
(0.5 mg/kg) three times weekly for 2 weeks, 66 participants (37 women) with a mean age of 36 years.
[195]
IFN-γ, IL-17α, IL-1β, IL-2, IL-23, IL-5, IL-6, IL-7, and TNF-α[195]
IL-8Depression, presence of a separate control group that received a placebo, depressed patients (n = 46, female, n = 17), cytokines were analyzed in plasma by multiplex bead-based immunoassays 24 h after infusion of ketamine (0.5 mg/kg).[226]
TNF-αTRD, randomized, double-blind control study, patients were randomized into three groups according to the treatment received: 0.5 mg/kg ketamine, 0.2 mg/kg ketamine, and normal saline infusion, cytokines were analyzed in plasma by ELISA test at baseline and at 40 min, 240 min, day 3, and day 7 post-infusion, 71 participants (53 women), with a mean age of 46 years.[246]
IL-6MDD, double-blind, placebo-controlled studies, cytokines were analyzed in plasma by multiplex bead-based immunoassays, blood samples were received at 60 min before ketamine infusion and at 230 min, one day, and three days post-infusion ketamine (0.5 mg/kg) or saline placebo, 80 participants (61 women), with a mean age of 45 years.[221]
TNF-αTRD, randomized placebo-controlled and open-label trials, cytokines were analyzed in plasma by ELISA test, 78 patients were allocated to receive two ketamine infusions (n = 30, days 1 and 4), a single ketamine (0.5 mg/kg) infusion (n = 24, only day 1), or normal saline placebo infusion (n = 24, only day 1). [247]
Esketamine IL-6 and CRPLabor and postpartum depression, a randomized, double-blinded controlled trial, cytokines were analyzed in plasma by ELISA test, a total of 120 women who underwent labor analgesia by epidural analgesia pump were enrolled and divided into two groups randomly. Esketamine at a dose of 0.2 mg/kg was intravenously injected after fetal disengagement in the test group and placebo was administered in the control group.[218]
IL-10[218]
IL-6Elective non-cardiac thoracic surgery under general anesthesia, randomized controlled trial.
During the operation, patients received 0.2 mg/kg (low-esketamine group) or 0.5 mg/kg esketamine (high-esketamine group) vs. placebo, cytokines were analyzed in plasma by ELISA test before surgery, post-operative day 1 and day 3, 129 participants (56 women), with a mean age of 65 years.
[219]

4. Discussion

Major depressive disorder (MDD) is the most common mental health condition worldwide, yet its underlying pathophysiology remains unclear. The monoamine hypothesis, which attributes depression to deficits in neurotransmitters like serotonin, norepinephrine, and dopamine, has traditionally shaped the development of antidepressants aimed at restoring monoamine balance in the synaptic cleft [14,23]. However, the prevalence of treatment-resistant depression and growing evidence from recent studies have led researchers to explore alternative mechanisms, including the potential role of inflammation and inflammatory markers in the development of depression [255,256,257]. These effects are mediated through multiple inflammatory pathways, including NF-κB, the NLRP3 inflammasome complex, the glutamatergic system, the gut–brain axis, the HPA axis impairing neuroplasticity, and the kynurenine pathway [74,75,76,77]. As this can be a very important avenue to be investigated in the pathophysiology of depression, we set out to combine all available human studies on the effects of antidepressants, specifically SSRIs, SNRIs, esketamine, and ketamine, on pro-inflammatory and anti-inflammatory markers, to see if there is a clear link between antidepressant treatment and a decrease in inflammation in the body.

4.1. Mechanisms of Anti-Inflammatory Action

The exact mechanism behind the anti-inflammatory effects of antidepressants remains unclear, though several theories have been proposed. It is known that ketamine exerts anti-inflammatory action through the NF-κB pathway [258]. Similarly, some evidence suggests that SSRIs and SNRIs may influence the NF-κB pathway and exert anti-inflammatory effects through the NLRP3 inflammasome complex [259].

4.1.1. NF-κB

The transcription factor NF-κB plays a central role in regulating both the innate and adaptive immune systems and acts as a key mediator of inflammation in the body [260]. It promotes the synthesis and release of pro-inflammatory cytokines [261] and has been found to be elevated in patients with depression [262]. There are two pathways of NF-κB activation in the CNS, the classical canonical pathway and the alternative non-canonical pathway [258], and they are activated by a variety of immune- and stress-related stimuli [263]. The canonical pathway is activated by various stimuli, including reactive oxygen species (ROS) [264], pathogen-associated molecular patterns (PAMPs), cytokines (TNF-α, TNF-β, IL-1), microbial components, and many other factors [263,265]. These triggers activate the IκB kinase complex (IKK), which phosphorylates the inhibitory IκB proteins, marking them for ubiquitination and subsequent degradation by the proteasome. This degradation releases the NF-κB dimers, allowing them to migrate into the nucleus and initiate the transcription of target genes [265,266,267,268].
Alternatively to the canonical pathway, the non-canonical pathway is only activated by specific stimuli, including the CD40 ligand, neurotrophic factors, lipopolysaccharide (LPS), and lymphotoxin β [267,269]. Furthermore, it does not rely on the IKK complex but instead is mediated by the NF-κB-inducing kinase (NIK), which activates IKKα [270,271,272]. This complex phosphorylates the NF-κB2 precursor protein, p100, marking it for ubiquitination and degradation by the proteasome. The resulting active complex translocates to the nucleus [267,269,270,271].
The NF-κB pathway has been suggested as a potential target for ketamine. Recent studies indicate that ketamine exerts a suppressive effect on NF-κB signaling in depression and inflammation models, leading to a reduction in levels of stress mediators and pro-inflammatory factors [258]. Reduced levels of NF-κB have been observed in various brain regions in animal studies [258] as well as in blood cells, further supporting this connection [273,274,275,276]. Furthermore, these studies have also shown that there is a potential link between NF-κB and BDNF, which could explain the anti-inflammatory and antidepressant effects of ketamine [258,277]. To date, only one clinical trial has investigated the effects of ketamine on NF-κB-mediated inflammation. Conducted in patients with acute lung injury induced by mechanical ventilation, the study demonstrated that ketamine administration significantly reduced serum levels of NF-κB and other inflammatory markers [278]. Ketamine inhibited NF-κB activity, as evidenced by decreased phosphorylation of the p65 subunit, and was associated with lower levels of oxidative stress markers and pro-inflammatory cytokines. These findings suggest that ketamine’s anti-inflammatory properties may involve suppression of NF-κB signaling [258].
Additionally, the modulation of the NF-κB signaling pathway by SSRIs and SNRIs has been investigated, with fluoxetine being the most extensively studied for its anti-inflammatory effects. Under ischemia/reperfusion conditions, fluoxetine decreased NF-κB activity by binding to IκB-α, thereby preventing its degradation and leading to increased IκB-α levels in BV-2 microglial cells [279]. Another study using a model of human hippocampal progenitor cells in an inflammatory environment demonstrated that treatment with venlafaxine and sertraline was associated with a decrease in NF-kB activity [280].

4.1.2. NLRP3 Inflammasome Complex

The Nod-like receptor protein 3 (NLRP3) inflammasome complex plays a central role in the innate immune response, providing protection against viral, bacterial, and fungal pathogens [281,282,283,284]. Once activated, the inflammasome can effectively mediate inflammation and initiate a robust immune response.
Recent studies have shown that activation of the NLRP3 inflammasome complex plays a role in the mediation of depression [259]. In mice subjected to chronic unpredictable mild stress (CUMS), which has been shown to yield the same effect as depression [285], levels of NLRP3 were increased in hippocampal regions [286,287]. Postmortem brain and serum analyses from patients with MDD have demonstrated activation of the NLRP3 inflammasome [288,289,290]. Supporting this observation, recent studies show that chronic antidepressant administration exerts anti-inflammatory effects, particularly within the hippocampus, suggesting that inhibition of NLRP3 inflammasome pathways may represent a potential mechanism underlying the antidepressant efficacy of drugs [259,291]. Ketamine has also been shown to exert antidepressant effects through inhibition of the NLRP3 inflammasome, as demonstrated in a recent study in a mouse model [292]. Furthermore, direct suppression of NLRP3 inflammasome activation by various inhibitors has been proposed as a promising therapeutic strategy for depression [293].

4.1.3. SSRIs

Due to the predominance of the monoamine theory of depression, SSRIs and SNRIs remain the most widely prescribed medications for the treatment of depression and are the first-line pharmacological interventions recommended in clinical guidelines [294]. Consequently, these drug classes have been the most extensively studied in relation to their effects on both inflammatory and non-inflammatory factors.

Fluoxetine

Fluoxetine was the first SSRI approved for the treatment of MDD in the US market [295,296]. Since its introduction, depression response rates have been between 60 and 70% with the various antidepressants that followed [297,298]. Fluoxetine is widely prescribed by physicians due to compelling characteristics such as low side effects, simple one-per-day dosing, and overdose safety. These factors contribute to better patient compliance and improved treatment outcomes [295].
Analyzing our results, we find that fluoxetine had the most studies on its effectiveness as an antidepressant. It was also found to primarily decrease the levels of most pro-inflammatory cytokines (IL-1β, IL-2, IL-6, IL-12, IL-15, NLRP3, IL-18, IFN-γ, TNF-α) in the studies [192,193,194,203,252,253] and increase the level of anti-inflammatory cytokines IL-4 [193,194] and IL-10 [231] (Table 1 and Figure 2). These results support our hypothesis that the administration of antidepressants can decrease pro-inflammatory factors and increase anti-inflammatory factors, which will overall result in a decrease in inflammation in the body.
Contradictory results to our hypothesis come from the same study, which found an increase in the pro-inflammatory factors IL-12 and IL-5 at week 8, and a decrease in the anti-inflammatory cytokine IL-1ra at week 4 which was then later restored at week 8, and no significant difference in clinical follow-up for anti-inflammatory cytokines IL-10 and IL-13; the reason for this is not explained by the authors [193]. Finally, two studies [196,205] found no significant effect on the pro-inflammatory factors IL-1β, IL-2, IL-6, and CRP (Table 1 and Figure 2).

Escitalopram

Escitalopram is characterized by being a potent and the most selective SSRI; it acts by specifically inhibiting SERT and consequently increasing serotonin levels in the CNS [299]. Pro-inflammatory factors (IL-1β, IL-1ra, IL-2, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-17, TNF-α, IFN-γ) were decreased across studies [191,203,215], with one study [198] noting no statistically significant effects on IL-10, IL-1ra, IL-6, IL-8, and TNF-α (Table 1 and Figure 2).
On the other hand, one study [191] showed that anti-inflammatory factors (IL-1ra, IL-10, IL-13) were decreased and pro-inflammatory factors (IL-12 and IL-15) were increased; this was not explained by the authors of the study (Table 1 and Figure 2).
A common pattern observed with the SSRIs fluoxetine and escitalopram is a decrease in the anti-inflammatory cytokine IL-1ra and an increase in the pro-inflammatory cytokine IL-12 following their administration.

Sertraline

Sertraline is a widely used SSRI that is effective for treating depression [300]. Furthermore, research has shown that sertraline outperforms other antidepressants, demonstrating greater efficacy than fluoxetine and better tolerability compared to amitriptyline, imipramine, paroxetine, and mirtazapine [300,301]. This could possibly explain why it has the most consistent results to support our hypothesis, as it has no contradictory increase in pro-inflammatory factors with the administration of sertraline. Pro-inflammatory factors (IL-2, IL-4, IL-6, IL-10, IL-17a, IFN-γ, TNF-α, and CRP) were decreased across studies [204,216,217], with three studies [204,222,223] noting no statistically significant effects on CRP, IL-6, and TNF-α. Additionally, an increase in the anti-inflammatory cytokine IL-10 was found in one study, although it was also not found to be statistically significant [216]. Moreover, a decrease in anti-inflammatory cytokines IL-4 and IL-10 was found in the same study [204] (Table 1 and Figure 2).

Paroxetine

Paroxetine is a well-tolerated SSRI with a low side effect profile and is effective in the treatment of MDD. It was also found to be effective in preventing relapses in patients treated for up to one year [302]. In the study investigated, an increase in the anti-inflammatory cytokine IL-10 and a contradictory increase in CRP were found [220]. A common pattern observed is an increase in the pro-inflammatory factors TNF-α and IL-6 in both paroxetine [220] and ketamine [221,247] (Table 1 and Figure 2). There are not enough current studies that follow our exclusion criteria to be able to draw any conclusions on the overall effect of paroxetine and fluvoxamine on anti-inflammation.

4.1.4. SNRIs: Venlafaxine

SNRIs were initially developed for patients with depression who did not respond to SSRIs, offering a treatment option with fewer side effects than tricyclic antidepressants. Studies have shown that at higher doses, venlafaxine is effective in treating resistant depression, with a side effect profile comparable to that of SSRIs [303]. Only CRP was studied in the article on venlafaxine, and its levels were decreased after treatment with it in patients with MDD [220] (Table 1 and Figure 2).

4.1.5. SSRIs vs. SNRIs

In conclusion, both SSRIs and SNRIs are found to support our hypothesis that antidepressants have an anti-inflammatory effect, and this is by decreasing levels of pro-inflammatory cytokines and increasing levels of anti-inflammatory factors. There are a few exceptions to some studies not following the same pattern and some showing no significant effects.

4.1.6. Ketamine/Esketamine

Ketamine has long been used as general anesthetic; however, recent studies have begun to explore its effectiveness in treating treatment-resistant depression [258,304,305,306]. Its mechanism of action primarily involves antagonism of the NMDA receptor and modulation of the NF-κB pathway, making its effects on depression and inflammation independent of the traditional monoamine hypothesis [258]. The use of ketamine for depression treatment remains a relatively new area of research, and its full therapeutic potential is still being investigated across various studies [304]. Pro-inflammatory factors (CRP, TNF-α, IL-6, IL-8/IL-10 ratio, IFN-γ, IL-17α, IL-1β, IL-2, IL-5, IL-6, IL-7) were decreased across studies [218,226,246,251,254]. Two studies investigating IL-6 and TNF-α separately showed them to increase upon administration of ketamine [221,247]. The authors of the former study concluded that post-infusion cytokine levels were not correlated with the antidepressant response to ketamine. Notably, the latter study observed an increase in TNF-α levels after two infusions of ketamine; alternatively, a single ketamine infusion was found to improve the TNF-α-to-IL-2 ratio when compared to either two ketamine infusions or a single placebo infusion [247]. Moreover, another study reported a decrease in anti-inflammatory cytokines IL-4 and IL-10 following ketamine administration [195].
Esketamine, a stereoisomer of ketamine, not only exhibits more potent anesthetic properties but has also shown effectiveness in treating individuals with severe, treatment-resistant depression [307]. It is currently the only NMDA receptor antagonist approved by the FDA and the European Medicines Agency (EMA) for this indication [42,307]. Both studies investigating esketamine reported a reduction in the pro-inflammatory cytokine IL-6, one in the context of labor and postpartum depression, and the other in patients undergoing elective non-cardiac thoracic surgery [218,219]. However, in the latter study, this effect was observed only with high-dose esketamine, as the low-dose regimen did not produce a significant change. Additionally, the former study also noted a decrease in C-reactive protein (CRP) levels and an increase in the anti-inflammatory cytokine IL-10, findings that are consistent with our hypothesis [218] (Table 1 and Figure 2). Given esketamine’s recent introduction into clinical practice, further research is warranted not only to thoroughly assess its long-term safety and therapeutic efficacy [307] but also to explore its potential immunomodulatory effects. Moreover, current studies investigating the relationship between ketamine and inflammation are limited; however, the existing evidence is highly promising and largely supportive of our hypothesis.

4.2. Limitations of Evidence

Comparing the results of these studies is challenging due to numerous limitations in measurement methods, reporting standards, and study settings. The studies reviewed exhibit considerable variability in sample sizes, ranging from as few as 22 participants to larger sample sizes of 73 [204], 90 [191], and 122 [223]. This variation introduces challenges in drawing generalizable conclusions. Additionally, the studies involve diverse age groups of adolescents [193], adults [191,204], and elderly people [223], which may further influence the outcomes and complicate cross-study comparisons. Differences in study duration also contribute to inconsistencies, with some studies spanning over 1 to 3 days [221], others extending to 4- and 8-week follow-ups [193], and some over 8-, 12-, and 26-week follow-ups [191].
Placebo-controlled trials are critical for ensuring the validity and interpretability of study outcomes. For example, one study reported a decrease in pro-inflammatory cytokines; however, similar decreases were observed in the placebo group [204]. This highlights the need for well-designed control conditions to accurately understand the effects of the treatment. Moreover, a common difference found across studies was the presence or absence of a control or placebo group. While the majority of studies included these groups [192,193,194,195,196,198,203,204,205,215,216,217,218,219,220,221,222,223,231,246,247,251,253,254,308], a few [191,226,252] did not, and instead measured only the changes over time within the same group of participants. This lack of a control group limits the ability to attribute observed changes to the intervention itself, thereby reducing the reproducibility and interpretive clarity of the findings, which can reduce the strength and reliability of the study’s conclusions. However, only 3 out of the 26 studies included in our review lacked a control group, representing a relatively small proportion of the total. Properly designed control and placebo groups are essential to ensure that observed effects can be confidently attributed to the intervention rather than to external or nonspecific influences. This enhances both the internal validity of the studies and the strength of their conclusions, thereby supporting more reliable scientific interpretations and future research directions in the field.
Another limitation of this review lies in the strict inclusion and exclusion criteria. Specifically, only studies that assessed inflammatory changes through cytokine measurement and simultaneously explored a connection with depression were included. While this approach ensured a focused analysis, it significantly reduced the number of eligible studies, potentially limiting the scope and generalizability of the findings. As a result, the conclusions drawn may not fully represent the wider body of existing literature on the subject, particularly studies employing alternative biomarkers or methodological frameworks.

5. Materials and Methods

5.1. Literature Search

A literature search was carried out in PubMed databases starting July 2024, using the queries ((escitalopram) or (esc))/(fluoxetine)/(Sertraline)/(Fluvoxamine)/(Paroxetine)/(Duloxetine)/(Venlafaxine)/(Ketamine)/(Esketamine) and (anti-inflammatory) or (inflammation) or (pro-inflammatory)) and ((depression) or (antidepressant)) Filters: Clinical Trial, Randomized Controlled Trial, from 2009 to 2024.
The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) flowchart is presented in Figure 3.

5.2. Eligibility Criteria

Not all studies identified through the search filter were included in this review. Despite the specificity of the search criteria, some retrieved studies did not meet the inclusion criteria, e.g., they did not examine depression as one of the conditions of the studies, or assess the impact of SSRIs, SNRIs, esketamine, or ketamine on inflammation; moreover, some studies were not conducted in vivo. Each search result was carefully screened, and only studies that explicitly addressed these key variables were selected. As a result, a significantly smaller number of studies were included after a thorough review of all retrieved articles.
A total of 114 studies were initially screened. Following the exclusion of duplicates and selection based on intervention, relevance, and study design, 27 studies met the eligibility criteria and were included in the analysis. Their results are discussed in the text and presented in Table 1.

6. Conclusions

The relationship between inflammation and depression is complex and remains an area of active investigation. Despite the numerous variables in the studies presented in this review, which are discussed in the limitations section, and the application of strict inclusion and exclusion criteria, it seems likely that there is a link between antidepressants and their anti-inflammatory effects. Levels of pro-inflammatory factors were mostly found to be decreased with treatment with SSRIs (escitalopram, fluoxetine, sertraline, paroxetine, and fluvoxamine), SNRIs (venlafaxine), esketamine, and ketamine. Additionally, the anti-inflammatory cytokine IL-10 was found to be increased upon treatment with fluoxetine, paroxetine, and esketamine, and IL-4 was increased upon treatment with fluoxetine. However, it should be emphasized that there are studies showing effects different from those described, which means that in this conclusion, we are only pointing out certain patterns and associations between antidepressant drug classes and changes in cytokine levels. This presents a new potential for investigating the pathophysiology of depression as it presents more evidence of its relationship with inflammation and how the treatment of inflammation could decrease depressive symptoms. Such findings can also provide insight into new therapeutic approaches for MDD. Further research is necessary to gain a more comprehensive understanding of the relationship between antidepressant therapy, inflammatory pathways, and treatment outcomes in depression. Long-term and larger-scale studies are needed to determine whether current findings reflect only acute effects or are sustained over time, and to evaluate the reproducibility and broader physiological impact of antidepressants. Additionally, (i) randomized controlled trials (RCTs) stratified by demographic factors such as age and gender are necessary to determine how these variables influence the inflammatory response and therapeutic outcomes. Incorporating pre-treatment and post-treatment inflammatory biomarker profiling would help elucidate differential responses across subgroups. Moreover, (ii) studies investigating specific molecular markers within inflammatory pathways, such as NF-κB and the NLRP3 inflammasome, may help identify predictors of therapeutic efficacy. Finally, given the limited existing evidence, (iii) further clinical trials exploring the anti-inflammatory and antidepressant effects of novel agents such as esketamine are also warranted.

Author Contributions

Conceptualization, A.W.-O. and L.B.; methodology, L.B., P.S. and A.W.-O.; writing—original draft preparation, L.B.; writing—review and editing, L.B., A.W.-O., P.S., G.H. and J.O.; visualization, L.B., A.W.-O. and J.O.; supervision, P.S. and A.W.-O.; funding acquisition, G.H., J.O. and A.W.-O. All authors have read and agreed to the published version of the manuscript.

Funding

This project has been financed by the Medical University of Lodz: 503/1-108-01/503-11-001 and 503/1-169-03/503-11-001 and 503/03-011-03/503-31-001.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. WHO Depressive Disorder (Depression). Available online: https://www.who.int/news-room/fact-sheets/detail/depression (accessed on 9 April 2024).
  2. Andrews, G.; Issakidis, C.; Sanderson, K.; Corry, J.; Lapsley, H. Utilising survey data to inform public policy: Comparison of the cost-effectiveness of treatment of ten mental disorders. Br. J. Psychiatry J. Ment. Sci. 2004, 184, 526–533. [Google Scholar] [CrossRef] [PubMed]
  3. Roose, S.P.; Schatzberg, A.F. The efficacy of antidepressants in the treatment of late-life depression. J. Clin. Psychopharmacol. 2005, 25, S1–S7. [Google Scholar] [CrossRef] [PubMed]
  4. Chisholm, D.; Sanderson, K.; Ayuso-Mateos, J.L.; Saxena, S. Reducing the global burden of depression: Population-level analysis of intervention cost-effectiveness in 14 world regions. Br. J. Psychiatry J. Ment. Sci. 2004, 184, 393–403. [Google Scholar] [CrossRef] [PubMed]
  5. Monroe, S.M.; Harkness, K.L. Recurrence in major depression: A conceptual analysis. Psychol. Rev. 2011, 118, 655–674. [Google Scholar] [CrossRef]
  6. Mathers, C.D.; Loncar, D. Projections of global mortality and burden of disease from 2002 to 2030. PLoS Med. 2006, 3, e442. [Google Scholar] [CrossRef]
  7. Institute for Health Metrics and Evaluation. IHME The Lancet: COVID-19 Pandemic Led to Stark Rise in Depressive and Anxiety Disorders Globally in 2020, with Women and Younger People Most Affected. Available online: https://www.healthdata.org/news-events/newsroom/news-releases/lancet-covid-19-pandemic-led-stark-rise-depressive-and-anxiety (accessed on 9 April 2024).
  8. Malhi, G.S.; Mann, J.J. Depression. Lancet 2018, 392, 2299–2312. [Google Scholar] [CrossRef]
  9. Stetler, C.; Miller, G.E. Depression and hypothalamic-pituitary-adrenal activation: A quantitative summary of four decades of research. Psychosom. Med. 2011, 73, 114–126. [Google Scholar] [CrossRef]
  10. Milaneschi, Y.; Lamers, F.; Berk, M.; Penninx, B.W.J.H. Depression Heterogeneity and Its Biological Underpinnings: Toward Immunometabolic Depression. Biol. Psychiatry 2020, 88, 369–380. [Google Scholar] [CrossRef]
  11. Cui, L.; Li, S.; Wang, S.; Wu, X.; Liu, Y.; Yu, W.; Wang, Y.; Tang, Y.; Xia, M.; Li, B. Major depressive disorder: Hypothesis, mechanism, prevention and treatment. Signal Transduct. Target. Ther. 2024, 9, 30. [Google Scholar] [CrossRef]
  12. Bunney, W.E.; Davis, J.M. Norepinephrine in depressive reactions. A review. Arch. Gen. Psychiatry 1965, 13, 483–494. [Google Scholar] [CrossRef]
  13. Schildkraut, J.J. The catecholamine hypothesis of affective disorders: A review of supporting evidence. Am. J. Psychiatry 1965, 122, 509–522. [Google Scholar] [CrossRef] [PubMed]
  14. Delgado, P.L. Depression: The case for a monoamine deficiency. J. Clin. Psychiatry 2000, 61 (Suppl. 6), 7–11. [Google Scholar] [PubMed]
  15. Hirschfeld, R.M.A. History and Evolution of the Monoamine Hypothesis of Depression. J. Clin. Psychiatry 2000, 61, 8272. [Google Scholar]
  16. Wong, D.T.; Horng, J.S.; Bymaster, F.P.; Hauser, K.L.; Molloy, B.B. A selective inhibitor of serotonin uptake: Lilly 110140, 3-(p-trifluoromethylphenoxy)-N-methyl-3-phenylpropylamine. Life Sci. 1974, 15, 471–479. [Google Scholar] [CrossRef]
  17. Wong, D.T.; Bymaster, F.P.; Engleman, E.A. Prozac (fluoxetine, Lilly 110140), the first selective serotonin uptake inhibitor and an antidepressant drug: Twenty years since its first publication. Life Sci. 1995, 57, 411–441. [Google Scholar] [CrossRef]
  18. Wong, D.T.; Perry, K.W.; Bymaster, F.P. Case history: The discovery of fluoxetine hydrochloride (Prozac). Nat. Rev. Drug Discov. 2005, 4, 764–774. [Google Scholar] [CrossRef]
  19. Papakostas, G.I. Serotonin Norepinephrine Reuptake Inhibitors: Spectrum of Efficacy in Major Depressive Disorder. Prim. Psychiatry 2009, 16, 16. [Google Scholar]
  20. Nestler, E.J.; Barrot, M.; DiLeone, R.J.; Eisch, A.J.; Gold, S.J.; Monteggia, L.M. Neurobiology of depression. Neuron 2002, 34, 13–25. [Google Scholar] [CrossRef]
  21. Hyman, S.E.; Nestler, E.J. Initiation and adaptation: A paradigm for understanding psychotropic drug action. Am. J. Psychiatry 1996, 153, 151–162. [Google Scholar] [CrossRef]
  22. Mill, J.; Petronis, A. Molecular studies of major depressive disorder: The epigenetic perspective. Mol. Psychiatry 2007, 12, 799–814. [Google Scholar] [CrossRef]
  23. Charney, D.S. Monoamine dysfunction and the pathophysiology and treatment of depression. J. Clin. Psychiatry 1998, 59 (Suppl. 14), 11–14. [Google Scholar] [PubMed]
  24. Heninger, G.R.; Delgado, P.L.; Charney, D.S. The revised monoamine theory of depression: A modulatory role for monoamines, based on new findings from monoamine depletion experiments in humans. Pharmacopsychiatry 1996, 29, 2–11. [Google Scholar] [CrossRef] [PubMed]
  25. Altamura, C.A.; Mauri, M.C.; Ferrara, A.; Moro, A.R.; D’Andrea, G.; Zamberlan, F. Plasma and platelet excitatory amino acids in psychiatric disorders. Am. J. Psychiatry 1993, 150, 1731–1733. [Google Scholar] [CrossRef]
  26. Küçükibrahimoğlu, E.; Saygin, M.Z.; Calişkan, M.; Kaplan, O.K.; Unsal, C.; Gören, M.Z. The change in plasma GABA, glutamine and glutamate levels in fluoxetine- or S-citalopram-treated female patients with major depression. Eur. J. Clin. Pharmacol. 2009, 65, 571–577. [Google Scholar] [CrossRef]
  27. Mauri, M.C.; Ferrara, A.; Boscati, L.; Bravin, S.; Zamberlan, F.; Alecci, M.; Invernizzi, G. Plasma and platelet amino acid concentrations in patients affected by major depression and under fluvoxamine treatment. Neuropsychobiology 1998, 37, 124–129. [Google Scholar] [CrossRef]
  28. Levine, J.; Panchalingam, K.; Rapoport, A.; Gershon, S.; McClure, R.J.; Pettegrew, J.W. Increased cerebrospinal fluid glutamine levels in depressed patients. Biol. Psychiatry 2000, 47, 586–593. [Google Scholar] [CrossRef]
  29. Domino, E.F. Taming the ketamine tiger. 1965. Anesthesiology 2010, 113, 678–684. [Google Scholar] [CrossRef]
  30. Lord, B.; Wintmolders, C.; Langlois, X.; Nguyen, L.; Lovenberg, T.; Bonaventure, P. Comparison of the ex vivo receptor occupancy profile of ketamine to several NMDA receptor antagonists in mouse hippocampus. Eur. J. Pharmacol. 2013, 715, 21–25. [Google Scholar] [CrossRef]
  31. Yamakura, T.; Shimoji, K. Subunit- and site-specific pharmacology of the NMDA receptor channel. Prog. Neurobiol. 1999, 59, 279–298. [Google Scholar] [CrossRef]
  32. Yamakura, T.; Mori, H.; Masaki, H.; Shimoji, K.; Mishina, M. Different sensitivities of NMDA receptor channel subtypes to non-competitive antagonists. Neuroreport 1993, 4, 687–690. [Google Scholar] [CrossRef]
  33. Mendelsohn, L.G.; Kalra, V.; Johnson, B.G.; Kerchner, G.A. Sigma opioid receptor: Characterization and co-identity with the phencyclidine receptor. J. Pharmacol. Exp. Ther. 1985, 233, 597–602. [Google Scholar] [CrossRef] [PubMed]
  34. Hirota, K.; Hashimoto, Y.; Lambert, D.G. Interaction of intravenous anesthetics with recombinant human M1-M3 muscarinic receptors expressed in chinese hamster ovary cells. Anesth. Analg. 2002, 95, 1607–1610. [Google Scholar] [CrossRef] [PubMed]
  35. Smith, D.J.; Bouchal, R.L.; deSanctis, C.A.; Monroe, P.J.; Amedro, J.B.; Perrotti, J.M.; Crisp, T. Properties of the interaction between ketamine and opiate binding sites in vivo and in vitro. Neuropharmacology 1987, 26, 1253–1260. [Google Scholar] [CrossRef]
  36. Nishimura, M.; Sato, K.; Okada, T.; Yoshiya, I.; Schloss, P.; Shimada, S.; Tohyama, M. Ketamine inhibits monoamine transporters expressed in human embryonic kidney 293 cells. Anesthesiology 1998, 88, 768–774. [Google Scholar] [CrossRef]
  37. Duman, R.S.; Li, N.; Liu, R.-J.; Duric, V.; Aghajanian, G. Signaling pathways underlying the rapid antidepressant actions of ketamine. Neuropharmacology 2012, 62, 35–41. [Google Scholar] [CrossRef] [PubMed]
  38. Berman, R.M.; Cappiello, A.; Anand, A.; Oren, D.A.; Heninger, G.R.; Charney, D.S.; Krystal, J.H. Antidepressant effects of ketamine in depressed patients. Biol. Psychiatry 2000, 47, 351–354. [Google Scholar] [CrossRef]
  39. Zarate, C.A.; Singh, J.B.; Carlson, P.J.; Brutsche, N.E.; Ameli, R.; Luckenbaugh, D.A.; Charney, D.S.; Manji, H.K. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch. Gen. Psychiatry 2006, 63, 856–864. [Google Scholar] [CrossRef]
  40. Daly, E.J.; Singh, J.B.; Fedgchin, M.; Cooper, K.; Lim, P.; Shelton, R.C.; Thase, M.E.; Winokur, A.; Van Nueten, L.; Manji, H.; et al. Efficacy and Safety of Intranasal Esketamine Adjunctive to Oral Antidepressant Therapy in Treatment-Resistant Depression: A Randomized Clinical Trial. JAMA Psychiatry 2018, 75, 139–148. [Google Scholar] [CrossRef]
  41. Daly, E.J.; Trivedi, M.H.; Janik, A.; Li, H.; Zhang, Y.; Li, X.; Lane, R.; Lim, P.; Duca, A.R.; Hough, D.; et al. Efficacy of Esketamine Nasal Spray Plus Oral Antidepressant Treatment for Relapse Prevention in Patients with Treatment-Resistant Depression: A Randomized Clinical Trial. JAMA Psychiatry 2019, 76, 893–903. [Google Scholar] [CrossRef]
  42. Cristea, I.A.; Naudet, F. US Food and Drug Administration approval of esketamine and brexanolone. Lancet Psychiatry 2019, 6, 975–977. [Google Scholar] [CrossRef]
  43. Lee, C.-H.; Giuliani, F. The Role of Inflammation in Depression and Fatigue. Front. Immunol. 2019, 10, 1696. [Google Scholar] [CrossRef] [PubMed]
  44. Qing, H.; Desrouleaux, R.; Israni-Winger, K.; Mineur, Y.S.; Fogelman, N.; Zhang, C.; Rashed, S.; Palm, N.W.; Sinha, R.; Picciotto, M.R.; et al. Origin and Function of Stress-Induced IL-6 in Murine Models. Cell 2020, 182, 372–387.e14. [Google Scholar] [CrossRef] [PubMed]
  45. Biltz, R.G.; Sawicki, C.M.; Sheridan, J.F.; Godbout, J.P. The neuroimmunology of social-stress-induced sensitization. Nat. Immunol. 2022, 23, 1527–1535. [Google Scholar] [CrossRef] [PubMed]
  46. Miller, A.H.; Maletic, V.; Raison, C.L. Inflammation and Its Discontents: The Role of Cytokines in the Pathophysiology of Major Depression. Biol. Psychiatry 2009, 65, 732–741. [Google Scholar] [CrossRef]
  47. Nadjar, A.; Bluthé, R.-M.; May, M.J.; Dantzer, R.; Parnet, P. Inactivation of the Cerebral NFκB Pathway Inhibits Interleukin-1β-Induced Sickness Behavior and c-Fos Expression in Various Brain Nuclei. Neuropsychopharmacology 2005, 30, 1492–1499. [Google Scholar] [CrossRef]
  48. Maes, M.; Bosmans, E.; De Jongh, R.; Kenis, G.; Vandoolaeghe, E.; Neels, H. Increased serum IL-6 and IL-1 receptor antagonist concentrations in major depression and treatment resistant depression. Cytokine 1997, 9, 853–858. [Google Scholar] [CrossRef]
  49. 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. [Google Scholar] [CrossRef]
  50. 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. [Google Scholar] [CrossRef]
  51. Varatharaj, A.; Galea, I. The blood-brain barrier in systemic inflammation. Brain. Behav. Immun. 2017, 60, 1–12. [Google Scholar] [CrossRef]
  52. Renault, P.F.; Hoofnagle, J.H.; Park, Y.; Mullen, K.D.; Peters, M.; Jones, D.B.; Rustgi, V.; Jones, E.A. Psychiatric complications of long-term interferon alfa therapy. Arch. Intern. Med. 1987, 147, 1577–1580. [Google Scholar] [CrossRef]
  53. Buter, J.; de Vries, E.G.; Sleijfer, D.T.; Willemse, P.H.; Mulder, N.H. Neuropsychiatric symptoms during treatment with interleukin-2. Lancet 1993, 341, 628. [Google Scholar] [CrossRef] [PubMed]
  54. Janssen, H.L.; Brouwer, J.T.; van der Mast, R.C.; Schalm, S.W. Suicide associated with alfa-interferon therapy for chronic viral hepatitis. J. Hepatol. 1994, 21, 241–243. [Google Scholar] [CrossRef] [PubMed]
  55. Capuron, L.; Ravaud, A.; Miller, A.H.; Dantzer, R. Baseline mood and psychosocial characteristics of patients developing depressive symptoms during interleukin-2 and/or interferon-alpha cancer therapy. Brain. Behav. Immun. 2004, 18, 205–213. [Google Scholar] [CrossRef] [PubMed]
  56. Dieperink, E.; Ho, S.B.; Tetrick, L.; Thuras, P.; Dua, K.; Willenbring, M.L. Suicidal ideation during interferon-alpha2b and ribavirin treatment of patients with chronic hepatitis C. Gen. Hosp. Psychiatry 2004, 26, 237–240. [Google Scholar] [CrossRef]
  57. Dunn, A.J.; Swiergiel, A.H.; de Beaurepaire, R. Cytokines as mediators of depression: What can we learn from animal studies? Neurosci. Biobehav. Rev. 2005, 29, 891–909. [Google Scholar] [CrossRef]
  58. Loftis, J.M.; Huckans, M.; Morasco, B.J. Neuroimmune mechanisms of cytokine-induced depression: Current theories and novel treatment strategies. Neurobiol. Dis. 2010, 37, 519–533. [Google Scholar] [CrossRef]
  59. Kohler, O.; Krogh, J.; Mors, O.; Benros, M.E. Inflammation in Depression and the Potential for Anti-Inflammatory Treatment. Curr. Neuropharmacol. 2016, 14, 732–742. [Google Scholar] [CrossRef]
  60. 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. [Google Scholar] [CrossRef]
  61. Hodes, G.E.; Kana, V.; Menard, C.; Merad, M.; Russo, S.J. Neuroimmune mechanisms of depression. Nat. Neurosci. 2015, 18, 1386–1393. [Google Scholar] [CrossRef]
  62. 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. [Google Scholar] [CrossRef]
  63. Jones, K.A.; Thomsen, C. The role of the innate immune system in psychiatric disorders. Mol. Cell. Neurosci. 2013, 53, 52–62. [Google Scholar] [CrossRef] [PubMed]
  64. Pace, T.W.W.; Miller, A.H. Cytokines and glucocorticoid receptor signaling. Relevance to major depression. Ann. N. Y. Acad. Sci. 2009, 1179, 86–105. [Google Scholar] [CrossRef] [PubMed]
  65. Söderlund, J.; Olsson, S.K.; Samuelsson, M.; Walther-Jallow, L.; Johansson, C.; Erhardt, S.; Landén, M.; Engberg, G. Elevation of cerebrospinal fluid interleukin-1β in bipolar disorder. J. Psychiatry Neurosci. JPN 2011, 36, 114–118. [Google Scholar] [CrossRef]
  66. Smith, K.J.; Au, B.; Ollis, L.; Schmitz, N. The association between C-reactive protein, Interleukin-6 and depression among older adults in the community: A systematic review and meta-analysis. Exp. Gerontol. 2018, 102, 109–132. [Google Scholar] [CrossRef]
  67. Korczak, D.J.; Pereira, S.; Koulajian, K.; Matejcek, A.; Giacca, A. Type 1 diabetes mellitus and major depressive disorder: Evidence for a biological link. Diabetologia 2011, 54, 2483–2493. [Google Scholar] [CrossRef]
  68. Dickens, C.; McGowan, L.; Clark-Carter, D.; Creed, F. Depression in rheumatoid arthritis: A systematic review of the literature with meta-analysis. Psychosom. Med. 2002, 64, 52–60. [Google Scholar] [CrossRef]
  69. You, Z.; Luo, C.; Zhang, W.; Chen, Y.; He, J.; Zhao, Q.; Zuo, R.; Wu, Y. Pro- and anti-inflammatory cytokines expression in rat’s brain and spleen exposed to chronic mild stress: Involvement in depression. Behav. Brain Res. 2011, 225, 135–141. [Google Scholar] [CrossRef]
  70. Osimo, E.F.; Baxter, L.J.; Lewis, G.; Jones, P.B.; Khandaker, G.M. Prevalence of low-grade inflammation in depression: A systematic review and meta-analysis of CRP levels. Psychol. Med. 2019, 49, 1958–1970. [Google Scholar] [CrossRef]
  71. Bialek, K.; Czarny, P.; Strycharz, J.; Sliwinski, T. Major depressive disorders accompanying autoimmune diseases—Response to treatment. Prog. Neuropsychopharmacol. Biol. Psychiatry 2019, 95, 109678. [Google Scholar] [CrossRef]
  72. Arteaga-Henríquez, G.; Simon, M.S.; Burger, B.; Weidinger, E.; Wijkhuijs, A.; Arolt, V.; Birkenhager, T.K.; Musil, R.; Müller, N.; Drexhage, H.A. Low-Grade Inflammation as a Predictor of Antidepressant and Anti-Inflammatory Therapy Response in MDD Patients: A Systematic Review of the Literature in Combination With an Analysis of Experimental Data Collected in the EU-MOODINFLAME Consortium. Front. Psychiatry 2019, 10, 458. [Google Scholar] [CrossRef]
  73. Jha, M.K.; Minhajuddin, A.; Gadad, B.S.; Greer, T.; Grannemann, B.; Soyombo, A.; Mayes, T.L.; Rush, A.J.; Trivedi, M.H. Can C-reactive protein inform antidepressant medication selection in depressed outpatients? Findings from the CO-MED trial. Psychoneuroendocrinology 2017, 78, 105–113. [Google Scholar] [CrossRef] [PubMed]
  74. Fries, G.R.; Saldana, V.A.; Finnstein, J.; Rein, T. Molecular pathways of major depressive disorder converge on the synapse. Mol. Psychiatry 2023, 28, 284–297. [Google Scholar] [CrossRef] [PubMed]
  75. Marx, W.; Penninx, B.W.J.H.; Solmi, M.; Furukawa, T.A.; Firth, J.; Carvalho, A.F.; Berk, M. Major depressive disorder. Nat. Rev. Dis. Primer 2023, 9, 44. [Google Scholar] [CrossRef]
  76. Patil, C.R.; Suryakant Gawli, C.; Bhatt, S. Targeting inflammatory pathways for treatment of the major depressive disorder. Drug Discov. Today 2023, 28, 103697. [Google Scholar] [CrossRef]
  77. Felger, J.C.; Lotrich, F.E. Inflammatory Cytokines in Depression: Neurobiological Mechanisms and Therapeutic Implications. Neuroscience 2013, 246, 199–229. [Google Scholar] [CrossRef]
  78. Al-Roub, A.; Al Madhoun, A.; Akhter, N.; Thomas, R.; Miranda, L.; Jacob, T.; Al-Ozairi, E.; Al-Mulla, F.; Sindhu, S.; Ahmad, R. IL-1β and TNFα Cooperativity in Regulating IL-6 Expression in Adipocytes Depends on CREB Binding and H3K14 Acetylation. Cells 2021, 10, 3228. [Google Scholar] [CrossRef]
  79. Chang, D.; Xing, Q.; Su, Y.; Zhao, X.; Xu, W.; Wang, X.; Dong, C. The Conserved Non-coding Sequences CNS6 and CNS9 Control Cytokine-Induced Rorc Transcription during T Helper 17 Cell Differentiation. Immunity 2020, 53, 614–626.e4. [Google Scholar] [CrossRef]
  80. Müller, N.; Ackenheil, M. Psychoneuroimmunology and the cytokine action in the CNS: Implications for psychiatric disorders. Prog. Neuropsychopharmacol. Biol. Psychiatry 1998, 22, 1–33. [Google Scholar] [CrossRef]
  81. 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. [Google Scholar] [CrossRef]
  82. Hong, H.; Kim, B.S.; Im, H.-I. Pathophysiological Role of Neuroinflammation in Neurodegenerative Diseases and Psychiatric Disorders. Int. Neurourol. J. 2016, 20, S2–S7. [Google Scholar] [CrossRef]
  83. Wohleb, E.S.; McKim, D.B.; Sheridan, J.F.; Godbout, J.P. Monocyte trafficking to the brain with stress and inflammation: A novel axis of immune-to-brain communication that influences mood and behavior. Front. Neurosci. 2014, 8, 447. [Google Scholar] [CrossRef] [PubMed]
  84. Najjar, S.; Pearlman, D.M.; Alper, K.; Najjar, A.; Devinsky, O. Neuroinflammation and psychiatric illness. J. Neuroinflamm. 2013, 10, 43. [Google Scholar] [CrossRef] [PubMed]
  85. Smith, R.S. The macrophage theory of depression. Med. Hypotheses 1991, 35, 298–306. [Google Scholar] [CrossRef] [PubMed]
  86. Wes, P.D.; Holtman, I.R.; Boddeke, E.W.G.M.; Möller, T.; Eggen, B.J.L. Next generation transcriptomics and genomics elucidate biological complexity of microglia in health and disease. Glia 2016, 64, 197–213. [Google Scholar] [CrossRef]
  87. Sousa, C.; Golebiewska, A.; Poovathingal, S.K.; Kaoma, T.; Pires-Afonso, Y.; Martina, S.; Coowar, D.; Azuaje, F.; Skupin, A.; Balling, R.; et al. Single-cell transcriptomics reveals distinct inflammation-induced microglia signatures. EMBO Rep. 2018, 19, e46171. [Google Scholar] [CrossRef]
  88. Lemaitre, P.; Tareen, S.H.; Pasciuto, E.; Mascali, L.; Martirosyan, A.; Callaerts-Vegh, Z.; Poovathingal, S.; Dooley, J.; Holt, M.G.; Yshii, L.; et al. Molecular and cognitive signatures of ageing partially restored through synthetic delivery of IL2 to the brain. EMBO Mol. Med. 2023, 15, e16805. [Google Scholar] [CrossRef]
  89. 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. [Google Scholar] [CrossRef]
  90. Zhou, Y.; Yan, M.; Pan, R.; Wang, Z.; Tao, X.; Li, C.; Xia, T.; Liu, X.; Chang, Q. Radix Polygalae extract exerts antidepressant effects in behavioral despair mice and chronic restraint stress-induced rats probably by promoting autophagy and inhibiting neuroinflammation. J. Ethnopharmacol. 2021, 265, 113317. [Google Scholar] [CrossRef]
  91. Qiu, X.; Wu, G.; Wang, L.; Tan, Y.; Song, Z. Lactobacillus delbrueckii alleviates depression-like behavior through inhibiting toll-like receptor 4 (TLR4) signaling in mice. Ann. Transl. Med. 2021, 9, 366. [Google Scholar] [CrossRef]
  92. He, Y.; Wang, Y.; Yu, H.; Tian, Y.; Chen, X.; Chen, C.; Ren, Y.; Chen, Z.; Ren, Y.; Gong, X.; et al. Protective effect of Nr4a2 (Nurr1) against LPS-induced depressive-like behaviors via regulating activity of microglia and CamkII neurons in anterior cingulate cortex. Pharmacol. Res. 2023, 191, 106717. [Google Scholar] [CrossRef]
  93. Li, H.; Xiang, Y.; Zhu, Z.; Wang, W.; Jiang, Z.; Zhao, M.; Cheng, S.; Pan, F.; Liu, D.; Ho, R.C.M.; et al. Rifaximin-mediated gut microbiota regulation modulates the function of microglia and protects against CUMS-induced depression-like behaviors in adolescent rat. J. Neuroinflamm. 2021, 18, 254. [Google Scholar] [CrossRef] [PubMed]
  94. Colonna, M.; Butovsky, O. Microglia Function in the Central Nervous System During Health and Neurodegeneration. Annu. Rev. Immunol. 2017, 35, 441–468. [Google Scholar] [CrossRef] [PubMed]
  95. Singhal, G.; Baune, B.T. Microglia: An Interface between the Loss of Neuroplasticity and Depression. Front. Cell. Neurosci. 2017, 11, 270. [Google Scholar] [CrossRef]
  96. Dwivedi, Y. Brain-derived neurotrophic factor: Role in depression and suicide. Neuropsychiatr. Dis. Treat. 2009, 5, 433–449. [Google Scholar] [CrossRef]
  97. Lee, B.-H.; Kim, Y.-K. The Roles of BDNF in the Pathophysiology of Major Depression and in Antidepressant Treatment. Psychiatry Investig. 2010, 7, 231–235. [Google Scholar] [CrossRef]
  98. Zhang, J.-C.; Yao, W.; Hashimoto, K. Brain-derived Neurotrophic Factor (BDNF)-TrkB Signaling in Inflammation-related Depression and Potential Therapeutic Targets. Curr. Neuropharmacol. 2016, 14, 721–731. [Google Scholar] [CrossRef]
  99. Lima Giacobbo, B.; Doorduin, J.; Klein, H.C.; Dierckx, R.A.J.O.; Bromberg, E.; de Vries, E.F.J. Brain-Derived Neurotrophic Factor in Brain Disorders: Focus on Neuroinflammation. Mol. Neurobiol. 2019, 56, 3295–3312. [Google Scholar] [CrossRef]
  100. Köhler-Forsberg, O.; Lydholm, C.N.; Hjorthøj, C.; Nordentoft, M.; Mors, O.; Benros, M.E. Efficacy of anti-inflammatory treatment on major depressive disorder or depressive symptoms: Meta-analysis of clinical trials. Acta Psychiatr. Scand. 2019, 139, 404–419. [Google Scholar] [CrossRef]
  101. Brites, D.; Fernandes, A. Neuroinflammation and Depression: Microglia Activation, Extracellular Microvesicles and microRNA Dysregulation. Front. Cell. Neurosci. 2015, 9, 476. [Google Scholar] [CrossRef]
  102. Liu, Z.; Fan, Y.; Won, S.J.; Neumann, M.; Hu, D.; Zhou, L.; Weinstein, P.R.; Liu, J. Chronic treatment with minocycline preserves adult new neurons and reduces functional impairment after focal cerebral ischemia. Stroke 2007, 38, 146–152. [Google Scholar] [CrossRef]
  103. Cabezas, R.; Avila-Rodriguez, M.; Vega-Vela, N.E.; Echeverria, V.; González, J.; Hidalgo, O.A.; Santos, A.B.; Aliev, G.; Barreto, G.E. Growth Factors and Astrocytes Metabolism: Possible Roles for Platelet Derived Growth Factor. Med. Chem. 2016, 12, 204–210. [Google Scholar] [CrossRef] [PubMed]
  104. Han, R.T.; Kim, R.D.; Molofsky, A.V.; Liddelow, S.A. Astrocyte-immune cell interactions in physiology and pathology. Immunity 2021, 54, 211–224. [Google Scholar] [CrossRef] [PubMed]
  105. Xu, S.; Xie, X.; Yao, L.; Wang, W.; Zhang, H.; Chen, M.; Sun, S.; Nie, Z.; Nagy, C.; Liu, Z. Human in vivo evidence of reduced astrocyte activation and neuroinflammation in patients with treatment-resistant depression following electroconvulsive therapy. Psychiatry Clin. Neurosci. 2023, 77, 653–664. [Google Scholar] [CrossRef] [PubMed]
  106. Carson, M.J.; Doose, J.M.; Melchior, B.; Schmid, C.D.; Ploix, C.C. CNS immune privilege: Hiding in plain sight. Immunol. Rev. 2006, 213, 48–65. [Google Scholar] [CrossRef]
  107. Schlaaff, K.; Dobrowolny, H.; Frodl, T.; Mawrin, C.; Gos, T.; Steiner, J.; Bogerts, B. Increased densities of T and B lymphocytes indicate neuroinflammation in subgroups of schizophrenia and mood disorder patients. Brain. Behav. Immun. 2020, 88, 497–506. [Google Scholar] [CrossRef]
  108. Wang, Y.-B.; Song, N.-N.; Ding, Y.-Q.; Zhang, L. Neural plasticity and depression treatment. IBRO Neurosci. Rep. 2023, 14, 160–184. [Google Scholar] [CrossRef]
  109. Fuchs, E.; Flügge, G. Adult Neuroplasticity: More Than 40 Years of Research. Neural Plast. 2014, 2014, 541870. [Google Scholar] [CrossRef]
  110. Toda, T.; Gage, F.H. Review: Adult neurogenesis contributes to hippocampal plasticity. Cell Tissue Res. 2018, 373, 693–709. [Google Scholar] [CrossRef]
  111. Albert, P.R. Adult neuroplasticity: A new “cure” for major depression? J. Psychiatry Neurosci. JPN 2019, 44, 147–150. [Google Scholar] [CrossRef]
  112. Liu, W.; Ge, T.; Leng, Y.; Pan, Z.; Fan, J.; Yang, W.; Cui, R. The Role of Neural Plasticity in Depression: From Hippocampus to Prefrontal Cortex. Neural Plast. 2017, 2017, 6871089. [Google Scholar] [CrossRef]
  113. Pittenger, C.; Duman, R.S. Stress, Depression, and Neuroplasticity: A Convergence of Mechanisms. Neuropsychopharmacology 2008, 33, 88–109. [Google Scholar] [CrossRef] [PubMed]
  114. Campbell, S.; Marriott, M.; Nahmias, C.; MacQueen, G.M. Lower hippocampal volume in patients suffering from depression: A meta-analysis. Am. J. Psychiatry 2004, 161, 598–607. [Google Scholar] [CrossRef] [PubMed]
  115. Chan, S.W.Y.; Harmer, C.J.; Norbury, R.; O’Sullivan, U.; Goodwin, G.M.; Portella, M.J. Hippocampal volume in vulnerability and resilience to depression. J. Affect. Disord. 2016, 189, 199–202. [Google Scholar] [CrossRef]
  116. Savitz, J.B.; Drevets, W.C. Imaging phenotypes of major depressive disorder: Genetic correlates. Neuroscience 2009, 164, 300–330. [Google Scholar] [CrossRef]
  117. Cobb, J.A.; Simpson, J.; Mahajan, G.J.; Overholser, J.C.; Jurjus, G.J.; Dieter, L.; Herbst, N.; May, W.; Rajkowska, G.; Stockmeier, C.A. Hippocampal volume and total cell numbers in major depressive disorder. J. Psychiatr. Res. 2013, 47, 299–306. [Google Scholar] [CrossRef]
  118. Rajkowska, G.; Miguel-Hidalgo, J.J.; Wei, J.; Dilley, G.; Pittman, S.D.; Meltzer, H.Y.; Overholser, J.C.; Roth, B.L.; Stockmeier, C.A. Morphometric evidence for neuronal and glial prefrontal cell pathology in major depression. Biol. Psychiatry 1999, 45, 1085–1098. [Google Scholar] [CrossRef]
  119. Duman, R.S.; Aghajanian, G.K. Synaptic dysfunction in depression: Potential therapeutic targets. Science 2012, 338, 68–72. [Google Scholar] [CrossRef]
  120. McEwen, B.S.; Eiland, L.; Hunter, R.G.; Miller, M.M. Stress and anxiety: Structural plasticity and epigenetic regulation as a consequence of stress. Neuropharmacology 2012, 62, 3–12. [Google Scholar] [CrossRef]
  121. Kuhn, M.; Höger, N.; Feige, B.; Blechert, J.; Normann, C.; Nissen, C. Fear Extinction as a Model for Synaptic Plasticity in Major Depressive Disorder. PLoS ONE 2014, 9, e115280. [Google Scholar] [CrossRef]
  122. Malberg, J.E.; Duman, R.S. Cell proliferation in adult hippocampus is decreased by inescapable stress: Reversal by fluoxetine treatment. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 2003, 28, 1562–1571. [Google Scholar] [CrossRef]
  123. Santarelli, L.; Saxe, M.; Gross, C.; Surget, A.; Battaglia, F.; Dulawa, S.; Weisstaub, N.; Lee, J.; Duman, R.; Arancio, O.; et al. Requirement of hippocampal neurogenesis for the behavioral effects of antidepressants. Science 2003, 301, 805–809. [Google Scholar] [CrossRef] [PubMed]
  124. Manji, H.K.; Moore, G.J.; Rajkowska, G.; Chen, G. Neuroplasticity and cellular resilience in mood disorders. Mol. Psychiatry 2000, 5, 578–593. [Google Scholar] [CrossRef] [PubMed]
  125. Magariños, A.M.; Deslandes, A.; McEwen, B.S. Effects of antidepressants and benzodiazepine treatments on the dendritic structure of CA3 pyramidal neurons after chronic stress. Eur. J. Pharmacol. 1999, 371, 113–122. [Google Scholar] [CrossRef] [PubMed]
  126. Boldrini, M.; Butt, T.H.; Santiago, A.N.; Tamir, H.; Dwork, A.J.; Rosoklija, G.B.; Arango, V.; Hen, R.; Mann, J.J. Benzodiazepines and the Potential Trophic Effect of Antidepressants on Dentate Gyrus Cells in Mood Disorders. Int. J. Neuropsychopharmacol. Off. Sci. J. Coll. Int. Neuropsychopharmacol. CINP 2014, 17, 1923–1933. [Google Scholar] [CrossRef]
  127. David, D.J.; Samuels, B.A.; Rainer, Q.; Wang, J.-W.; Marsteller, D.; Mendez, I.; Drew, M.; Craig, D.A.; Guiard, B.P.; Guilloux, J.-P.; et al. Neurogenesis-Dependent and -Independent Effects of Fluoxetine in an Animal Model of Anxiety/Depression. Neuron 2009, 62, 479–493. [Google Scholar] [CrossRef]
  128. Andreyev, A.Y.; Kushnareva, Y.E.; Starkov, A.A. Mitochondrial metabolism of reactive oxygen species. Biochem. Biokhimiia 2005, 70, 200–214. [Google Scholar] [CrossRef]
  129. Bajpai, A. Oxidative Stress and Major Depression. J. Clin. Diagn. Res. 2014, 8, CC04–CC07. [Google Scholar] [CrossRef]
  130. Ďuračková, Z. Some current insights into oxidative stress. Physiol. Res. 2010, 59, 459–469. [Google Scholar] [CrossRef]
  131. Popa-Wagner, A.; Mitran, S.; Sivanesan, S.; Chang, E.; Buga, A.-M. ROS and Brain Diseases: The Good, the Bad, and the Ugly. Oxid. Med. Cell. Longev. 2013, 2013, 963520. [Google Scholar] [CrossRef]
  132. McCracken, E.; Valeriani, V.; Simpson, C.; Jover, T.; McCulloch, J.; Dewar, D. The lipid peroxidation by-product 4-hydroxynonenal is toxic to axons and oligodendrocytes. J. Cereb. Blood Flow Metab. 2000, 20, 1529–1536. [Google Scholar] [CrossRef]
  133. Correia, A.S.; Cardoso, A.; Vale, N. Oxidative Stress in Depression: The Link with the Stress Response, Neuroinflammation, Serotonin, Neurogenesis and Synaptic Plasticity. Antioxidants 2023, 12, 470. [Google Scholar] [CrossRef] [PubMed]
  134. Bhatt, S.; Nagappa, A.N.; Patil, C.R. Role of oxidative stress in depression. Drug Discov. Today 2020, 25, 1270–1276. [Google Scholar] [CrossRef] [PubMed]
  135. Lindqvist, D.; Dhabhar, F.S.; James, S.J.; Hough, C.M.; Jain, F.A.; Bersani, F.S.; Reus, V.I.; Verhoeven, J.E.; Epel, E.S.; Mahan, L.; et al. Oxidative stress, inflammation and treatment response in major depression. Psychoneuroendocrinology 2017, 76, 197–205. [Google Scholar] [CrossRef] [PubMed]
  136. Maes, M.; Galecki, P.; Chang, Y.S.; Berk, M. A review on the oxidative and nitrosative stress (O&NS) pathways in major depression and their possible contribution to the (neuro)degenerative processes in that illness. Prog. Neuropsychopharmacol. Biol. Psychiatry 2011, 35, 676–692. [Google Scholar] [CrossRef]
  137. Michel, T.M.; Frangou, S.; Thiemeyer, D.; Camara, S.; Jecel, J.; Nara, K.; Brunklaus, A.; Zoechling, R.; Riederer, P. Evidence for oxidative stress in the frontal cortex in patients with recurrent depressive disorder--a postmortem study. Psychiatry Res. 2007, 151, 145–150. [Google Scholar] [CrossRef]
  138. Michel, T.M.; Thome, J.; Martin, D.; Nara, K.; Zwerina, S.; Tatschner, T.; Weijers, H.G.; Koutsilieri, E. Cu, Zn- and Mn-superoxide dismutase levels in brains of patients with schizophrenic psychosis. J. Neural Transm. 2004, 111, 1191–1201. [Google Scholar] [CrossRef]
  139. Michel, T.M.; Sybille, C.; Thomas, T.; Sophia, F.; Jane, S.A.; Peter, R.; Grünblatt, E. Increased xanthine oxidase in the thalamus and putamen in depression. World J. Biol. Psychiatry 2010, 11, 314–320. [Google Scholar] [CrossRef]
  140. Bouayed, J.; Rammal, H.; Soulimani, R. Oxidative Stress and Anxiety: Relationship and Cellular Pathways. Oxid. Med. Cell. Longev. 2009, 2, 623654. [Google Scholar] [CrossRef]
  141. Mahmoud, A.M.; Alexander, M.Y.; Tutar, Y.; Wilkinson, F.L.; Venditti, A. Oxidative Stress in Metabolic Disorders and Drug-Induced Injury: The Potential Role of Nrf2 and PPARs Activators. Oxid. Med. Cell. Longev. 2017, 2017, 2508909. [Google Scholar] [CrossRef]
  142. Murphy, B.E. Steroids and depression. J. Steroid Biochem. Mol. Biol. 1991, 38, 537–559. [Google Scholar] [CrossRef]
  143. Anders, S.; Tanaka, M.; Kinney, D.K. Depression as an evolutionary strategy for defense against infection. Brain. Behav. Immun. 2013, 31, 9–22. [Google Scholar] [CrossRef]
  144. Milne, A.M.B.; MacQueen, G.M.; Hall, G.B.C. Abnormal hippocampal activation in patients with extensive history of major depression: An fMRI study. J. Psychiatry Neurosci. JPN 2012, 37, 28–36. [Google Scholar] [CrossRef] [PubMed]
  145. Stewart, J.G.; Mazurka, R.; Bond, L.; Wynne-Edwards, K.E.; Harkness, K.L. Rumination and impaired cortisol recovery following a social stressor in adolescent depression. J. Abnorm. Child Psychol. 2013, 41, 1015–1026. [Google Scholar] [CrossRef] [PubMed]
  146. Nikkheslat, N.; Zunszain, P.A.; Horowitz, M.A.; Barbosa, I.G.; Parker, J.A.; Myint, A.-M.; Schwarz, M.J.; Tylee, A.T.; Carvalho, L.A.; Pariante, C.M. Insufficient glucocorticoid signaling and elevated inflammation in coronary heart disease patients with comorbid depression. Brain. Behav. Immun. 2015, 48, 8–18. [Google Scholar] [CrossRef]
  147. Kokras, N.; Hodes, G.E.; Bangasser, D.A.; Dalla, C. Sex differences in the hypothalamic–pituitary–adrenal axis: An obstacle to antidepressant drug development? Br. J. Pharmacol. 2019, 176, 4090–4106. [Google Scholar] [CrossRef]
  148. Frank, M.G.; Hershman, S.A.; Weber, M.D.; Watkins, L.R.; Maier, S.F. Chronic exposure to exogenous glucocorticoids primes microglia to pro-inflammatory stimuli and induces NLRP3 mRNA in the hippocampus. Psychoneuroendocrinology 2014, 40, 191–200. [Google Scholar] [CrossRef]
  149. Chai, Y.; Cai, Y.; Fu, Y.; Wang, Y.; Zhang, Y.; Zhang, X.; Zhu, L.; Miao, M.; Yan, T. Salidroside Ameliorates Depression by Suppressing NLRP3-Mediated Pyroptosis via P2X7/NF-κB/NLRP3 Signaling Pathway. Front. Pharmacol. 2022, 13, 812362. [Google Scholar] [CrossRef]
  150. Sapolsky, R.M.; Krey, L.C.; McEwen, B.S. Prolonged glucocorticoid exposure reduces hippocampal neuron number: Implications for aging. J. Neurosci. Off. J. Soc. Neurosci. 1985, 5, 1222–1227. [Google Scholar] [CrossRef]
  151. Zhang, Z.; Deng, T.; Wu, M.; Zhu, A.; Zhu, G. Botanicals as modulators of depression and mechanisms involved. Chin. Med. 2019, 14, 24. [Google Scholar] [CrossRef]
  152. Wong, M.L.; Lewis, M.; Licinio, J. Translational research in endocrinology and neuroimmunology applied to depression. In Biomedical Chemistry; De Gruyter: Berlin, Germany, 2015; pp. 119–131. ISBN 978-3-11-046874-8. [Google Scholar] [CrossRef]
  153. Badawy, A.A.-B. Kynurenine Pathway of Tryptophan Metabolism: Regulatory and Functional Aspects. Int. J. Tryptophan Res. IJTR 2017, 10, 1178646917691938. [Google Scholar] [CrossRef]
  154. Agudelo, L.Z.; Femenía, T.; Orhan, F.; Porsmyr-Palmertz, M.; Goiny, M.; Martinez-Redondo, V.; Correia, J.C.; Izadi, M.; Bhat, M.; Schuppe-Koistinen, I.; et al. Skeletal Muscle PGC-1α1 Modulates Kynurenine Metabolism and Mediates Resilience to Stress-Induced Depression. Cell 2014, 159, 33–45. [Google Scholar] [CrossRef] [PubMed]
  155. Sforzini, L.; Pariante, C.M.; Palacios, J.E.; Tylee, A.; Carvalho, L.A.; Viganò, C.A.; Nikkheslat, N. Inflammation associated with coronary heart disease predicts onset of depression in a three-year prospective follow-up: A preliminary study. Brain. Behav. Immun. 2019, 81, 659–664. [Google Scholar] [CrossRef] [PubMed]
  156. Hunt, C.; Macedo e Cordeiro, T.; Suchting, R.; de Dios, C.; Cuellar Leal, V.A.; Soares, J.C.; Dantzer, R.; Teixeira, A.L.; Selvaraj, S. Effect of immune activation on the kynurenine pathway and depression symptoms—A systematic review and meta-analysis. Neurosci. Biobehav. Rev. 2020, 118, 514–523. [Google Scholar] [CrossRef]
  157. Dantzer, R. Role of the kynurenine metabolism pathway in inflammation-induced depression—Preclinical approaches. Curr. Top. Behav. Neurosci. 2017, 31, 117–138. [Google Scholar] [CrossRef]
  158. Vécsei, L.; Szalárdy, L.; Fülöp, F.; Toldi, J. Kynurenines in the CNS: Recent advances and new questions. Nat. Rev. Drug Discov. 2013, 12, 64–82. [Google Scholar] [CrossRef]
  159. Mándi, Y.; Stone, T.W.; Guillemin, G.J.; Vécsei, L.; Williams, R.O. Editorial: Multiple Implications of the Kynurenine Pathway in Inflammatory Diseases: Diagnostic and Therapeutic Applications. Front. Immunol. 2022, 13, 860867. [Google Scholar] [CrossRef]
  160. Bay-Richter, C.; Linderholm, K.R.; Lim, C.K.; Samuelsson, M.; Träskman-Bendz, L.; Guillemin, G.J.; Erhardt, S.; Brundin, L. A role for inflammatory metabolites as modulators of the glutamate N-methyl-D-aspartate receptor in depression and suicidality. Brain. Behav. Immun. 2015, 43, 110–117. [Google Scholar] [CrossRef]
  161. Carabotti, M.; Scirocco, A.; Maselli, M.A.; Severi, C. The gut-brain axis: Interactions between enteric microbiota, central and enteric nervous systems. Ann. Gastroenterol. Q. Publ. Hell. Soc. Gastroenterol. 2015, 28, 203–209. [Google Scholar]
  162. Rhee, S.H.; Pothoulakis, C.; Mayer, E.A. Principles and clinical implications of the brain-gut-enteric microbiota axis. Nat. Rev. Gastroenterol. Hepatol. 2009, 6, 306–314. [Google Scholar] [CrossRef]
  163. Frankiensztajn, L.M.; Elliott, E.; Koren, O. The microbiota and the hypothalamus-pituitary-adrenocortical (HPA) axis, implications for anxiety and stress disorders. Curr. Opin. Neurobiol. 2020, 62, 76–82. [Google Scholar] [CrossRef]
  164. Mohammadi, G.; Dargahi, L.; Peymani, A.; Mirzanejad, Y.; Alizadeh, S.A.; Naserpour, T.; Nassiri-Asl, M. The Effects of Probiotic Formulation Pretreatment (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) on a Lipopolysaccharide Rat Model. J. Am. Coll. Nutr. 2019, 38, 209–217. [Google Scholar] [CrossRef] [PubMed]
  165. Duvallet, C.; Gibbons, S.M.; Gurry, T.; Irizarry, R.A.; Alm, E.J. Meta-analysis of gut microbiome studies identifies disease-specific and shared responses. Nat. Commun. 2017, 8, 1784. [Google Scholar] [CrossRef] [PubMed]
  166. Burberry, A.; Wells, M.F.; Limone, F.; Couto, A.; Smith, K.S.; Keaney, J.; Gillet, G.; van Gastel, N.; Wang, J.-Y.; Pietilainen, O.; et al. C9orf72 suppresses systemic and neural inflammation induced by gut bacteria. Nature 2020, 582, 89–94. [Google Scholar] [CrossRef] [PubMed]
  167. Blacher, E.; Bashiardes, S.; Shapiro, H.; Rothschild, D.; Mor, U.; Dori-Bachash, M.; Kleimeyer, C.; Moresi, C.; Harnik, Y.; Zur, M.; et al. Potential roles of gut microbiome and metabolites in modulating ALS in mice. Nature 2019, 572, 474–480. [Google Scholar] [CrossRef]
  168. Tillmann, S.; Awwad, H.M.; Eskelund, A.R.; Treccani, G.; Geisel, J.; Wegener, G.; Obeid, R. Probiotics Affect One-Carbon Metabolites and Catecholamines in a Genetic Rat Model of Depression. Mol. Nutr. Food Res. 2018, 62, e1701070. [Google Scholar] [CrossRef]
  169. Chen, Y.; Xu, J.; Chen, Y. Regulation of Neurotransmitters by the Gut Microbiota and Effects on Cognition in Neurological Disorders. Nutrients 2021, 13, 2099. [Google Scholar] [CrossRef]
  170. Karbownik, M.S.; Sokołowska, P.; Kowalczyk, E. Gut Microbiota Metabolites Differentially Release Gliotransmitters from the Cultured Human Astrocytes: A Preliminary Report. Int. J. Mol. Sci. 2023, 24, 6617. [Google Scholar] [CrossRef]
  171. Bleibel, L.; Dziomba, S.; Waleron, K.F.; Kowalczyk, E.; Karbownik, M.S. Deciphering psychobiotics’ mechanism of action: Bacterial extracellular vesicles in the spotlight. Front. Microbiol. 2023, 14, 1211447. [Google Scholar] [CrossRef]
  172. Rathour, D.; Shah, S.; Khan, S.; Singh, P.K.; Srivastava, S.; Singh, S.B.; Khatri, D.K. Role of gut microbiota in depression: Understanding molecular pathways, recent research, and future direction. Behav. Brain Res. 2023, 436, 114081. [Google Scholar] [CrossRef]
  173. Hao, W.; Ma, Q.; Wang, L.; Yuan, N.; Gan, H.; He, L.; Li, X.; Huang, J.; Chen, J. Gut dysbiosis induces the development of depression-like behavior through abnormal synapse pruning in microglia-mediated by complement C3. Microbiome 2024, 12, 34. [Google Scholar] [CrossRef]
  174. Rudzki, L.; Maes, M. The Microbiota-Gut-Immune-Glia (MGIG) Axis in Major Depression. Mol. Neurobiol. 2020, 57, 4269–4295. [Google Scholar] [CrossRef] [PubMed]
  175. Roy Sarkar, S.; Banerjee, S. Gut microbiota in neurodegenerative disorders. J. Neuroimmunol. 2019, 328, 98–104. [Google Scholar] [CrossRef] [PubMed]
  176. Maes, M. The cytokine hypothesis of depression: Inflammation, oxidative & nitrosative stress (IO&NS) and leaky gut as new targets for adjunctive treatments in depression. Neuro Endocrinol. Lett. 2008, 29, 287–291. [Google Scholar] [PubMed]
  177. Bisgaard, T.H.; Allin, K.H.; Keefer, L.; Ananthakrishnan, A.N.; Jess, T. Depression and anxiety in inflammatory bowel disease: Epidemiology, mechanisms and treatment. Nat. Rev. Gastroenterol. Hepatol. 2022, 19, 717–726. [Google Scholar] [CrossRef]
  178. Spiller, R.; Garsed, K. Postinfectious irritable bowel syndrome. Gastroenterology 2009, 136, 1979–1988. [Google Scholar] [CrossRef]
  179. Clouse, R.E.; Lustman, P.J.; Geisman, R.A.; Alpers, D.H. Antidepressant therapy in 138 patients with irritable bowel syndrome: A five-year clinical experience. Aliment. Pharmacol. Ther. 1994, 8, 409–416. [Google Scholar] [CrossRef]
  180. Amirani, E.; Milajerdi, A.; Mirzaei, H.; Jamilian, H.; Mansournia, M.A.; Hallajzadeh, J.; Ghaderi, A. The effects of probiotic supplementation on mental health, biomarkers of inflammation and oxidative stress in patients with psychiatric disorders: A systematic review and meta-analysis of randomized controlled trials. Complement. Ther. Med. 2020, 49, 102361. [Google Scholar] [CrossRef]
  181. Messaoudi, M.; Lalonde, R.; Violle, N.; Javelot, H.; Desor, D.; Nejdi, A.; Bisson, J.-F.; Rougeot, C.; Pichelin, M.; Cazaubiel, M.; et al. Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. Br. J. Nutr. 2011, 105, 755–764. [Google Scholar] [CrossRef]
  182. Huang, R.; Ning, H.; Yang, L.; Jia, C.; Yang, F.; Xu, G.; Tan, H. Efficacy of Probiotics on Anxiety: A Meta-analysis of Randomized Controlled Trials. Neuropsychiatry 2017, 7, 862–871. [Google Scholar] [CrossRef]
  183. Reis, D.J.; Ilardi, S.S.; Punt, S.E.W. The anxiolytic effect of probiotics: A systematic review and meta-analysis of the clinical and preclinical literature. PLoS ONE 2018, 13, e0199041. [Google Scholar] [CrossRef]
  184. Goh, K.K.; Liu, Y.-W.; Kuo, P.-H.; Chung, Y.-C.E.; Lu, M.-L.; Chen, C.-H. Effect of probiotics on depressive symptoms: A meta-analysis of human studies. Psychiatry Res. 2019, 282, 112568. [Google Scholar] [CrossRef] [PubMed]
  185. Chao, L.; Liu, C.; Sutthawongwadee, S.; Li, Y.; Lv, W.; Chen, W.; Yu, L.; Zhou, J.; Guo, A.; Li, Z.; et al. Effects of Probiotics on Depressive or Anxiety Variables in Healthy Participants Under Stress Conditions or With a Depressive or Anxiety Diagnosis: A Meta-Analysis of Randomized Controlled Trials. Front. Neurol. 2020, 11, 421. [Google Scholar] [CrossRef] [PubMed]
  186. Cohen Kadosh, K.; Basso, M.; Knytl, P.; Johnstone, N.; Lau, J.Y.F.; Gibson, G.R. Psychobiotic interventions for anxiety in young people: A systematic review and meta-analysis, with youth consultation. Transl. Psychiatry 2021, 11, 352. [Google Scholar] [CrossRef]
  187. El Dib, R.; Periyasamy, A.G.; de Barros, J.L.; França, C.G.; Senefonte, F.L.; Vesentini, G.; Alves, M.G.O.; Rodrigues, J.V.d.S.; Gomaa, H.; Gomes Júnior, J.R.; et al. Probiotics for the treatment of depression and anxiety: A systematic review and meta-analysis of randomized controlled trials. Clin. Nutr. ESPEN 2021, 45, 75–90. [Google Scholar] [CrossRef]
  188. Liu, R.T.; Walsh, R.F.L.; Sheehan, A.E. Prebiotics and probiotics for depression and anxiety: A systematic review and meta-analysis of controlled clinical trials. Neurosci. Biobehav. Rev. 2019, 102, 13–23. [Google Scholar] [CrossRef]
  189. Dinarello, C.A. Biologic Basis for Interleukin-1 in Disease. Blood 1996, 87, 2095–2147. [Google Scholar] [CrossRef]
  190. Zou, W.; Feng, R.; Yang, Y. Changes in the serum levels of inflammatory cytokines in antidepressant drug-naïve patients with major depression. PLoS ONE 2018, 13, e0197267. [Google Scholar] [CrossRef]
  191. Kofod, J.; Elfving, B.; Nielsen, E.H.; Mors, O.; Köhler-Forsberg, O. Depression and inflammation: Correlation between changes in inflammatory markers with antidepressant response and long-term prognosis. Eur. Neuropsychopharmacol. 2022, 54, 116–125. [Google Scholar] [CrossRef]
  192. Song, C.; Halbreich, U.; Han, C.; Leonard, B.E.; Luo, H. Imbalance between pro- and anti-inflammatory cytokines, and between Th1 and Th2 cytokines in depressed patients: The effect of electroacupuncture or fluoxetine treatment. Pharmacopsychiatry 2009, 42, 182–188. [Google Scholar] [CrossRef]
  193. Pérez-Sánchez, G.; Becerril-Villanueva, E.; Arreola, R.; Martínez-Levy, G.; Hernández-Gutiérrez, M.E.; Velasco-Velásquez, M.A.; Alvarez-Herrera, S.; Cruz-Fuentes, C.; Palacios, L.; de la Peña, F.; et al. Inflammatory Profiles in Depressed Adolescents Treated with Fluoxetine: An 8-Week Follow-up Open Study. Mediators Inflamm. 2018, 2018, 4074051. [Google Scholar] [CrossRef]
  194. Guo, J.; Zhang, T.; Chen, W.; Tan, J.; Li, X.; Zheng, A.; Fu, Y.; Qiu, T. The relationship between serum resolvin D1, NLRP3, cytokine levels, and adolescents with first-episode medication-naïve major depressive disorder. BMC Psychiatry 2024, 24, 285. [Google Scholar] [CrossRef] [PubMed]
  195. Zhou, Y.; Wang, C.; Lan, X.; Li, H.; Chao, Z.; Ning, Y. Plasma inflammatory cytokines and treatment-resistant depression with comorbid pain: Improvement by ketamine. J. Neuroinflamm. 2021, 18, 200. [Google Scholar] [CrossRef] [PubMed]
  196. Jazayeri, S.; Keshavarz, S.A.; Tehrani-Doost, M.; Djalali, M.; Hosseini, M.; Amini, H.; Chamari, M.; Djazayery, A. Effects of eicosapentaenoic acid and fluoxetine on plasma cortisol, serum interleukin-1beta and interleukin-6 concentrations in patients with major depressive disorder. Psychiatry Res. 2010, 178, 112–115. [Google Scholar] [CrossRef]
  197. Dinarello, C.A.; Thompson, R.C. Blocking IL-1: Interleukin 1 receptor antagonist in vivo and in vitro. Immunol. Today 1991, 12, 404–410. [Google Scholar] [CrossRef]
  198. Haastrup, E.; Knorr, U.; Erikstrup, C.; Kessing, L.V.; Ullum, H. No evidence for an anti-inflammatory effect of escitalopram intervention in healthy individuals with a family history of depression. J. Neuroimmunol. 2012, 243, 69–72. [Google Scholar] [CrossRef]
  199. Kalia, V.; Sarkar, S.; Subramaniam, S.; Haining, W.N.; Smith, K.A.; Ahmed, R. Prolonged interleukin-2Ralpha expression on virus-specific CD8+ T cells favors terminal-effector differentiation in vivo. Immunity 2010, 32, 91–103. [Google Scholar] [CrossRef]
  200. Pipkin, M.E.; Sacks, J.A.; Cruz-Guilloty, F.; Lichtenheld, M.G.; Bevan, M.J.; Rao, A. Interleukin-2 and inflammation induce distinct transcriptional programs that promote the differentiation of effector cytolytic T cells. Immunity 2010, 32, 79–90. [Google Scholar] [CrossRef]
  201. Williams, M.A.; Tyznik, A.J.; Bevan, M.J. Interleukin-2 signals during priming are required for secondary expansion of CD8+ memory T cells. Nature 2006, 441, 890–893. [Google Scholar] [CrossRef]
  202. Malek, T.R.; Castro, I. Interleukin-2 receptor signaling: At the interface between tolerance and immunity. Immunity 2010, 33, 153–165. [Google Scholar] [CrossRef]
  203. Xiaoling, Z.; Yunping, H.; Yingdong, L. Analysis of curative effect of fluoxetine and escitalopram in the depression treatment based on clinical observation. Pak. J. Pharm. Sci. 2018, 31, 1115–1118. [Google Scholar]
  204. Brunoni, A.R.; Machado-Vieira, R.; Zarate, C.A.; Valiengo, L.; Vieira, E.L.; Benseñor, I.M.; Lotufo, P.A.; Gattaz, W.F.; Teixeira, A.L. Cytokines plasma levels during antidepressant treatment with sertraline and transcranial direct current stimulation (tDCS): Results from a factorial, randomized, controlled trial. Psychopharmacology 2014, 231, 1315–1323. [Google Scholar] [CrossRef] [PubMed]
  205. Mackay, G.M.; Forrest, C.M.; Christofides, J.; Bridel, M.A.; Mitchell, S.; Cowlard, R.; Stone, T.W.; Darlington, L.G. Kynurenine metabolites and inflammation markers in depressed patients treated with fluoxetine or counselling. Clin. Exp. Pharmacol. Physiol. 2009, 36, 425–435. [Google Scholar] [CrossRef] [PubMed]
  206. Brown, M.A.; Hural, J. Functions of IL-4 and control of its expression. Crit. Rev. Immunol. 1997, 17, 1–32. [Google Scholar] [CrossRef] [PubMed]
  207. Hart, P.H.; Vitti, G.F.; Burgess, D.R.; Whitty, G.A.; Piccoli, D.S.; Hamilton, J.A. Potential antiinflammatory effects of interleukin 4: Suppression of human monocyte tumor necrosis factor alpha, interleukin 1, and prostaglandin E2. Proc. Natl. Acad. Sci. USA 1989, 86, 3803–3807. [Google Scholar] [CrossRef]
  208. Fenton, M.J.; Buras, J.A.; Donnelly, R.P. IL-4 reciprocally regulates IL-1 and IL-1 receptor antagonist expression in human monocytes. J. Immunol. 1992, 149, 1283–1288. [Google Scholar] [CrossRef]
  209. Greenfeder, S.; Umland, S.P.; Cuss, F.M.; Chapman, R.W.; Egan, R.W. Th2 cytokines and asthma—The role of interleukin-5 in allergic eosinophilic disease. Respir. Res. 2001, 2, 71. [Google Scholar] [CrossRef]
  210. Pelaia, C.; Paoletti, G.; Puggioni, F.; Racca, F.; Pelaia, G.; Canonica, G.W.; Heffler, E. Interleukin-5 in the Pathophysiology of Severe Asthma. Front. Physiol. 2019, 10, 1514. [Google Scholar] [CrossRef]
  211. Korn, T.; Bettelli, E.; Oukka, M.; Kuchroo, V.K. IL-17 and Th17 Cells. Annu. Rev. Immunol. 2009, 27, 485–517. [Google Scholar] [CrossRef]
  212. Kumar, H.; Kawai, T.; Akira, S. Pathogen recognition by the innate immune system. Int. Rev. Immunol. 2011, 30, 16–34. [Google Scholar] [CrossRef]
  213. Heinrich, P.C.; Castell, J.V.; Andus, T. Interleukin-6 and the acute phase response. Biochem. J. 1990, 265, 621–636. [Google Scholar] [CrossRef]
  214. Kishimoto, T. Factors affecting B-cell growth and differentiation. Annu. Rev. Immunol. 1985, 3, 133–157. [Google Scholar] [CrossRef] [PubMed]
  215. Cattaneo, A.; Gennarelli, M.; Uher, R.; Breen, G.; Farmer, A.; Aitchison, K.J.; Craig, I.W.; Anacker, C.; Zunsztain, P.A.; McGuffin, P.; et al. Candidate Genes Expression Profile Associated with Antidepressants Response in the GENDEP Study: Differentiating between Baseline ‘Predictors’ and Longitudinal ‘Targets’. Neuropsychopharmacology 2013, 38, 377–385. [Google Scholar] [CrossRef]
  216. Taraz, M.; Khatami, M.-R.; Dashti-Khavidaki, S.; Akhonzadeh, S.; Noorbala, A.-A.; Ghaeli, P.; Taraz, S. Sertraline decreases serum level of interleukin-6 (IL-6) in hemodialysis patients with depression: Results of a randomized double-blind, placebo-controlled clinical trial. Int. Immunopharmacol. 2013, 17, 917–923. [Google Scholar] [CrossRef]
  217. Pizzi, C.; Mancini, S.; Angeloni, L.; Fontana, F.; Manzoli, L.; Costa, G. Effects of Selective Serotonin Reuptake Inhibitor Therapy on Endothelial Function and Inflammatory Markers in Patients with Coronary Heart Disease. Clin. Pharmacol. Ther. 2009, 86, 527–532. [Google Scholar] [CrossRef]
  218. Ling, B.; Zhu, Y.; Yan, Z.; Chen, H.; Xu, H.; Wang, Q.; Yu, W.; Wang, W. Effect of single intravenous injection of esketamine on postpartum depression after labor analgesia and potential mechanisms: A randomized, double-blinded controlled trial. BMC Pharmacol. Toxicol. 2023, 24, 66. [Google Scholar] [CrossRef]
  219. Luo, T.; Deng, Z.; Ren, Q.; Mu, F.; Zhang, Y.; Wang, H. Effects of esketamine on postoperative negative emotions and early cognitive disorders in patients undergoing non-cardiac thoracic surgery: A randomized controlled trial. J. Clin. Anesth. 2024, 95, 111447. [Google Scholar] [CrossRef]
  220. Carboni, L.; McCarthy, D.J.; Delafont, B.; Filosi, M.; Ivanchenko, E.; Ratti, E.; Learned, S.M.; Alexander, R.; Domenici, E. Biomarkers for response in major depression: Comparing paroxetine and venlafaxine from two randomised placebo-controlled clinical studies. Transl. Psychiatry 2019, 9, 182. [Google Scholar] [CrossRef]
  221. Park, M.; Newman, L.E.; Gold, P.W.; Luckenbaugh, D.A.; Yuan, P.; Machado-Vieira, R.; Zarate, C.A. Change in Cytokine Levels is Not Associated with Rapid Antidepressant Response to Ketamine in Treatment-Resistant Depression. J. Psychiatr. Res. 2017, 84, 113–118. [Google Scholar] [CrossRef]
  222. Gregg, L.P.; Carmody, T.; Le, D.; Bharadwaj, N.; Trivedi, M.H.; Hedayati, S.S. Depression and the Effect of Sertraline on Inflammatory Biomarkers in Patients with Nondialysis CKD. Kidney360 2020, 1, 436–446. [Google Scholar] [CrossRef]
  223. Bot, M.; Carney, R.M.; Freedland, K.E.; Rubin, E.H.; Rich, M.W.; Steinmeyer, B.C.; Mann, D.L. Inflammation and treatment response to sertraline in patients with coronary heart disease and comorbid major depression. J. Psychosom. Res. 2011, 71, 13–17. [Google Scholar] [CrossRef]
  224. Chen, D.; Tang, T.-X.; Deng, H.; Yang, X.-P.; Tang, Z.-H. Interleukin-7 Biology and Its Effects on Immune Cells: Mediator of Generation, Differentiation, Survival, and Homeostasis. Front. Immunol. 2021, 12, 747324. [Google Scholar] [CrossRef] [PubMed]
  225. Bickel, M. The role of interleukin-8 in inflammation and mechanisms of regulation. J. Periodontol. 1993, 64, 456–460. [Google Scholar] [PubMed]
  226. 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. [Google Scholar] [CrossRef] [PubMed]
  227. O’Shea, J.J.; Gadina, M.; Siegel, R. 9—Cytokines and cytokine receptors. In Clinical Immunology, 4th ed.; Rich, R.R., Fleisher, T.A., Shearer, W.T., Schroeder, H.W., Frew, A.J., Weyand, C.M., Eds.; Elsevier: London, UK, 2013; pp. 108–135. ISBN 978-0-7234-3691-1. [Google Scholar]
  228. Rojas-Zuleta, W.G.; Sanchez, E. IL-9: Function, Sources, and Detection. In Th9 Cells: Methods and Protocols; Goswami, R., Ed.; Springer: New York, NY, USA, 2017; pp. 21–35. ISBN 978-1-4939-6877-0. [Google Scholar]
  229. Sabat, R.; Grütz, G.; Warszawska, K.; Kirsch, S.; Witte, E.; Wolk, K.; Geginat, J. Biology of interleukin-10. Cytokine Growth Factor Rev. 2010, 21, 331–344. [Google Scholar] [CrossRef]
  230. Kühn, R.; Löhler, J.; Rennick, D.; Rajewsky, K.; Müller, W. Interleukin-10-deficient mice develop chronic enterocolitis. Cell 1993, 75, 263–274. [Google Scholar] [CrossRef]
  231. Mikocka-Walus, A.; Hughes, P.A.; Bampton, P.; Gordon, A.; Campaniello, M.A.; Mavrangelos, C.; Stewart, B.J.; Esterman, A.; Andrews, J.M. Fluoxetine for Maintenance of Remission and to Improve Quality of Life in Patients with Crohn’s Disease: A Pilot Randomized Placebo-Controlled Trial. J. Crohns Colitis 2016, 11, 509. [Google Scholar] [CrossRef]
  232. Horowitz, M.C.; Lorenzo, J.A. Chapter 57—Local Regulators of Bone: IL-1, TNF, Lymphotoxin, Interferon-γ, the LIF/IL-6 Family, and Additional Cytokines. In Principles of Bone Biology, 3rd ed.; Bilezikian, J.P., Raisz, L.G., Martin, T.J., Eds.; Academic Press: San Diego, CA, USA, 2008; pp. 1209–1234. ISBN 978-0-12-373884-4. [Google Scholar]
  233. Aste-Amezaga, M.; D’Andrea, A.; Kubin, M.; Trinchieri, G. Cooperation of natural killer cell stimulatory factor/interleukin-12 with other stimuli in the induction of cytokines and cytotoxic cell-associated molecules in human T and NK cells. Cell. Immunol. 1994, 156, 480–492. [Google Scholar] [CrossRef]
  234. Perussia, B.; Chan, S.H.; D’Andrea, A.; Tsuji, K.; Santoli, D.; Pospisil, M.; Young, D.; Wolf, S.F.; Trinchieri, G. Natural killer (NK) cell stimulatory factor or IL-12 has differential effects on the proliferation of TCR-alpha beta+, TCR-gamma delta+ T lymphocytes, and NK cells. J. Immunol. 1992, 149, 3495–3502. [Google Scholar] [CrossRef]
  235. Salcedo, T.W.; Azzoni, L.; Wolf, S.F.; Perussia, B. Modulation of perforin and granzyme messenger RNA expression in human natural killer cells. J. Immunol. 1993, 151, 2511–2520. [Google Scholar] [CrossRef]
  236. Trinchieri, G. Interleukin-12: A proinflammatory cytokine with immunoregulatory functions that bridge innate resistance and antigen-specific adaptive immunity. Annu. Rev. Immunol. 1995, 13, 251–276. [Google Scholar] [CrossRef]
  237. Wynn, T.A. IL-13 effector functions. Annu. Rev. Immunol. 2003, 21, 425–456. [Google Scholar] [CrossRef] [PubMed]
  238. de Vries, J.E. The role of IL-13 and its receptor in allergy and inflammatory responses. J. Allergy Clin. Immunol. 1998, 102, 165–169. [Google Scholar] [CrossRef] [PubMed]
  239. Perera, P.-Y.; Lichy, J.H.; Waldmann, T.A.; Perera, L.P. The role of Interleukin-15 in inflammation and immune responses to infection: Implications for its therapeutic use. Microbes Infect. Inst. Pasteur 2011, 14, 247. [Google Scholar] [CrossRef] [PubMed]
  240. Kolls, J.K.; Lindén, A. Interleukin-17 family members and inflammation. Immunity 2004, 21, 467–476. [Google Scholar] [CrossRef]
  241. Billiau, A. Interferon-gamma: Biology and role in pathogenesis. Adv. Immunol. 1996, 62, 61–130. [Google Scholar] [CrossRef]
  242. Boehm, U.; Klamp, T.; Groot, M.; Howard, J.C. Cellular responses to interferon-gamma. Annu. Rev. Immunol. 1997, 15, 749–795. [Google Scholar] [CrossRef]
  243. Idriss, H.T.; Naismith, J.H. TNFα and the TNF receptor superfamily: Structure-function relationship(s). Microsc. Res. Tech. 2000, 50, 184–195. [Google Scholar] [CrossRef]
  244. Beyaert, R.; Fiers, W. Molecular mechanisms of tumor necrosis factor-induced cytotoxicity. What we do understand and what we do not. FEBS Lett. 1994, 340, 9–16. [Google Scholar] [CrossRef]
  245. Bradley, J.R. TNF-mediated inflammatory disease. J. Pathol. 2008, 214, 149–160. [Google Scholar] [CrossRef]
  246. Chen, M.-H.; Li, C.-T.; Lin, W.-C.; Hong, C.-J.; Tu, P.-C.; Bai, Y.-M.; Cheng, C.-M.; Su, T.-P. Rapid inflammation modulation and antidepressant efficacy of a low-dose ketamine infusion in treatment-resistant depression: A randomized, double-blind control study. Psychiatry Res. 2018, 269, 207–211. [Google Scholar] [CrossRef]
  247. Chen, M.-H.; Wu, H.-J.; Li, C.-T.; Lin, W.-C.; Tsai, S.-J.; Hong, C.-J.; Tu, P.-C.; Bai, Y.-M.; Mao, W.-C.; Su, T.-P. Is one or two infusions better in the first week of low-dose ketamine treatment for medication-resistant depression? A post hoc pooled analysis of randomized placebo-controlled and open-label trials. J. Psychiatr. Res. 2021, 144, 448–454. [Google Scholar] [CrossRef] [PubMed]
  248. Yano, T. 3—The Nonspecific Immune System: Humoral Defense. In Fish Physiology; Iwama, G., Nakanishi, T., Eds.; Organism, Pathogen, and Environment; Academic Press: Cambridge, MA, USA, 1996; Volume 15, pp. 105–157. [Google Scholar]
  249. Du Clos, T.W.; Mold, C. C-reactive protein: An activator of innate immunity and a modulator of adaptive immunity. Immunol. Res. 2004, 30, 261–277. [Google Scholar] [CrossRef] [PubMed]
  250. Du Clos, T.W. Function of C-reactive protein. Ann. Med. 2000, 32, 274–278. [Google Scholar] [CrossRef] [PubMed]
  251. Hudetz, J.A.; Iqbal, Z.; Gandhi, S.D.; Patterson, K.M.; Byrne, A.J.; Hudetz, A.G.; Pagel, P.S.; Warltier, D.C. Ketamine attenuates post-operative cognitive dysfunction after cardiac surgery. Acta Anaesthesiol. Scand. 2009, 53, 864–872. [Google Scholar] [CrossRef]
  252. Gupta, K.; Gupta, R.; Bhatia, M.S.; Tripathi, A.K.; Gupta, L.K. Effect of Agomelatine and Fluoxetine on HAM-D Score, Serum Brain-Derived Neurotrophic Factor, and Tumor Necrosis Factor-α Level in Patients with Major Depressive Disorder with Severe Depression. J. Clin. Pharmacol. 2017, 57, 1519–1526. [Google Scholar] [CrossRef]
  253. Sedighi, F.; Zarghami, M.; Alizadeh Arimi, F.; Moosazadeh, M.; Ala, S.; Ghasemian, R.; Mehravaran, H.; Elyasi, F. Efficacy and safety of adding fluoxetine to the treatment regimen of hospitalized patients with non-critical COVID-19 pneumonia: A double-blind randomized, placebo-controlled clinical trial. Neuropsychopharmacol. Rep. 2023, 43, 202–212. [Google Scholar] [CrossRef]
  254. Langhein, M.; Seitz-Holland, J.; Lyall, A.E.; Pasternak, O.; Chunga, N.; Cetin-Karayumak, S.; Kubicki, A.; Mulert, C.; Espinoza, R.T.; Narr, K.L.; et al. Association between peripheral inflammation and free-water imaging in Major Depressive Disorder before and after ketamine treatment—A pilot study. J. Affect. Disord. 2022, 314, 78–85. [Google Scholar] [CrossRef]
  255. Haroon, E.; Daguanno, A.W.; Woolwine, B.J.; Goldsmith, D.R.; Baer, W.M.; Wommack, E.C.; Felger, J.C.; Miller, A.H. Antidepressant treatment resistance is associated with increased inflammatory markers in patients with major depressive disorder. Psychoneuroendocrinology 2018, 95, 43–49. [Google Scholar] [CrossRef]
  256. Hamon, M.; Blier, P. Monoamine neurocircuitry in depression and strategies for new treatments. Prog. Neuropsychopharmacol. Biol. Psychiatry 2013, 45, 54–63. [Google Scholar] [CrossRef]
  257. Caldiroli, A.; Capuzzi, E.; Tagliabue, I.; Capellazzi, M.; Marcatili, M.; Mucci, F.; Colmegna, F.; Clerici, M.; Buoli, M.; Dakanalis, A. Augmentative Pharmacological Strategies in Treatment-Resistant Major Depression: A Comprehensive Review. Int. J. Mol. Sci. 2021, 22, 13070. [Google Scholar] [CrossRef]
  258. Sokołowska, P.; Seweryn Karbownik, M.; Jóźwiak-Bębenista, M.; Dobielska, M.; Kowalczyk, E.; Wiktorowska-Owczarek, A. Antidepressant mechanisms of ketamine’s action: NF-κB in the spotlight. Biochem. Pharmacol. 2023, 218, 115918. [Google Scholar] [CrossRef] [PubMed]
  259. Xia, C.-Y.; Guo, Y.-X.; Lian, W.-W.; Yan, Y.; Ma, B.-Z.; Cheng, Y.-C.; Xu, J.-K.; He, J.; Zhang, W.-K. The NLRP3 inflammasome in depression: Potential mechanisms and therapies. Pharmacol. Res. 2023, 187, 106625. [Google Scholar] [CrossRef] [PubMed]
  260. Liu, T.; Zhang, L.; Joo, D.; Sun, S.-C. NF-κB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef]
  261. Tak, P.P.; Firestein, G.S. NF-κB: A key role in inflammatory diseases. J. Clin. Investig. 2001, 107, 7–11. [Google Scholar] [CrossRef]
  262. Pace, T.W.W.; Mletzko, T.C.; Alagbe, O.; Musselman, D.L.; Nemeroff, C.B.; Miller, A.H.; Heim, C.M. Increased Stress-Induced Inflammatory Responses in Male Patients with Major Depression and Increased Early Life Stress. Am. J. Psychiatry 2006, 163, 1630–1633. [Google Scholar] [CrossRef]
  263. Zhang, H.; Sun, S.-C. NF-κB in inflammation and renal diseases. Cell Biosci. 2015, 5, 63. [Google Scholar] [CrossRef]
  264. Gloire, G.; Legrand-Poels, S.; Piette, J. NF-κB activation by reactive oxygen species: Fifteen years later. Biochem. Pharmacol. 2006, 72, 1493–1505. [Google Scholar] [CrossRef]
  265. Mincheva-Tasheva, S.; Soler, R.M. NF-κB Signaling Pathways: Role in Nervous System Physiology and Pathology. Neuroscientist 2013, 19, 175–194. [Google Scholar] [CrossRef]
  266. Napetschnig, J.; Wu, H. Molecular Basis of NF-κB Signaling. Annu. Rev. Biophys. 2013, 42, 443–468. [Google Scholar] [CrossRef]
  267. Oeckinghaus, A.; Hayden, M.S.; Ghosh, S. Crosstalk in NF-κB signaling pathways. Nat. Immunol. 2011, 12, 695–708. [Google Scholar] [CrossRef]
  268. Karin, M. The Beginning of the End: IκB Kinase (IKK) and NF-κB Activation. J. Biol. Chem. 1999, 274, 27339–27342. [Google Scholar] [CrossRef] [PubMed]
  269. Hayden, M.S.; Ghosh, S. NF-κB in immunobiology. Cell Res. 2011, 21, 223–244. [Google Scholar] [CrossRef] [PubMed]
  270. Senftleben, U.; Cao, Y.; Xiao, G.; Greten, F.R.; Krähn, G.; Bonizzi, G.; Chen, Y.; Hu, Y.; Fong, A.; Sun, S.C.; et al. Activation by IKKalpha of a second, evolutionary conserved, NF-kappa B signaling pathway. Science 2001, 293, 1495–1499. [Google Scholar] [CrossRef] [PubMed]
  271. Xiao, G.; Harhaj, E.W.; Sun, S.C. NF-kappaB-inducing kinase regulates the processing of NF-kappaB2 p100. Mol. Cell 2001, 7, 401–409. [Google Scholar] [CrossRef]
  272. Sun, S.-C. Non-canonical NF-κB signaling pathway. Cell Res. 2011, 21, 71–85. [Google Scholar] [CrossRef]
  273. Yu, Y.; Zhou, Z.; Xu, J.; Liu, Z.; Wang, Y. Ketamine reduces NFkappaB activation and TNFalpha production in rat mononuclear cells induced by lipopolysaccharide in vitro. Ann. Clin. Lab. Sci. 2002, 32, 292–298. [Google Scholar]
  274. Sun, J.; Zhou, Z.Q.; Lv, R.; Li, W.Y.; Xu, J.G. Ketamine inhibits LPS-induced calcium elevation and NF-kappa B activation in monocytes. Inflamm. Res. 2004, 53, 304–308. [Google Scholar] [CrossRef]
  275. Chang, H.-C.; Lin, K.-H.; Tai, Y.-T.; Chen, J.-T.; Chen, R.-M. Lipoteichoic acid-induced TNF-α and IL-6 gene expressions and oxidative stress production in macrophages are suppressed by ketamine through downregulating Toll-like receptor 2-mediated activation oF ERK1/2 and NFκB. Shock Augusta Ga 2010, 33, 485–492. [Google Scholar] [CrossRef]
  276. Welters, I.D.; Hafer, G.; Menzebach, A.; Mühling, J.; Neuhäuser, C.; Browning, P.; Goumon, Y. Ketamine Inhibits Transcription Factors Activator Protein 1 and Nuclear Factor-κB, Interleukin-8 Production, as well as CD11b and CD16 Expression: Studies in Human Leukocytes and Leukocytic Cell Lines. Anesth. Analg. 2010, 110, 934. [Google Scholar] [CrossRef]
  277. Caviedes, A.; Lafourcade, C.; Soto, C.; Wyneken, U. BDNF/NF-κB Signaling in the Neurobiology of Depression. Curr. Pharm. Des. 2017, 23, 3154–3163. [Google Scholar] [CrossRef]
  278. Wang, W.-F.; Liu, S.; Xu, B. A study of the protective effect and mechanism of ketamine on acute lung injury induced by mechanical ventilation. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 1362–1367. [Google Scholar] [PubMed]
  279. Tian, M.; Yang, M.; Li, Z.; Wang, Y.; Chen, W.; Yang, L.; Li, Y.; Yuan, H. Fluoxetine suppresses inflammatory reaction in microglia under OGD/R challenge via modulation of NF-κB signaling. Biosci. Rep. 2019, 39, BSR20181584. [Google Scholar] [CrossRef] [PubMed]
  280. Horowitz, M.A.; Wertz, J.; Zhu, D.; Cattaneo, A.; Musaelyan, K.; Nikkheslat, N.; Thuret, S.; Pariante, C.M.; Zunszain, P.A. Antidepressant Compounds Can Be Both Pro- and Anti-Inflammatory in Human Hippocampal Cells. Int. J. Neuropsychopharmacol. 2015, 18, pyu076. [Google Scholar] [CrossRef]
  281. Thomas, P.G.; Dash, P.; Aldridge, J.R.; Ellebedy, A.H.; Reynolds, C.; Funk, A.J.; Martin, W.J.; Lamkanfi, M.; Webby, R.J.; Boyd, K.L.; et al. The intracellular sensor NLRP3 mediates key innate and healing responses to influenza A virus via the regulation of caspase-1. Immunity 2009, 30, 566–575. [Google Scholar] [CrossRef]
  282. Allen, I.C.; Scull, M.A.; Moore, C.B.; Holl, E.K.; McElvania-TeKippe, E.; Taxman, D.J.; Guthrie, E.H.; Pickles, R.J.; Ting, J.P.-Y. The NLRP3 inflammasome mediates in vivo innate immunity to influenza A virus through recognition of viral RNA. Immunity 2009, 30, 556–565. [Google Scholar] [CrossRef]
  283. Gross, O.; Poeck, H.; Bscheider, M.; Dostert, C.; Hannesschläger, N.; Endres, S.; Hartmann, G.; Tardivel, A.; Schweighoffer, E.; Tybulewicz, V.; et al. Syk kinase signalling couples to the Nlrp3 inflammasome for anti-fungal host defence. Nature 2009, 459, 433–436. [Google Scholar] [CrossRef]
  284. Kanneganti, T.-D.; Body-Malapel, M.; Amer, A.; Park, J.-H.; Whitfield, J.; Franchi, L.; Taraporewala, Z.F.; Miller, D.; Patton, J.T.; Inohara, N.; et al. Critical role for Cryopyrin/Nalp3 in activation of caspase-1 in response to viral infection and double-stranded RNA. J. Biol. Chem. 2006, 281, 36560–36568. [Google Scholar] [CrossRef]
  285. Antoniuk, S.; Bijata, M.; Ponimaskin, E.; Wlodarczyk, J. Chronic unpredictable mild stress for modeling depression in rodents: Meta-analysis of model reliability. Neurosci. Biobehav. Rev. 2019, 99, 101–116. [Google Scholar] [CrossRef]
  286. Chen, H.; Ma, Y.; Chen, M.; Chen, J.; Chen, J. Safflower extract improves depression in mice by inhibiting the TLR4-NLRP3 inflammation signaling pathway. Ann. Palliat. Med. 2021, 10, 8015023–8018023. [Google Scholar] [CrossRef]
  287. Sahin, C.; Albayrak, O.; Akdeniz, T.F.; Akbulut, Z.; Yanikkaya Demirel, G.; Aricioglu, F. Agmatine Reverses Sub-chronic Stress induced Nod-like Receptor Protein 3 (NLRP3) Activation and Cytokine Response in Rats. Basic Clin. Pharmacol. Toxicol. 2016, 119, 367–375. [Google Scholar] [CrossRef]
  288. Pandey, G.N.; Zhang, H.; Sharma, A.; Ren, X. Innate immunity receptors in depression and suicide: Upregulated NOD-like receptors containing pyrin (NLRPs) and hyperactive inflammasomes in the postmortem brains of people who were depressed and died by suicide. J. Psychiatry Neurosci. 2021, 46, E538–E547. [Google Scholar] [CrossRef] [PubMed]
  289. Taene, A.; Khalili-Tanha, G.; Esmaeili, A.; Mobasheri, L.; Kooshkaki, O.; Jafari, S.; Shokouhifar, A.; Sarab, G.A. The Association of Major Depressive Disorder with Activation of NLRP3 Inflammasome, Lipid Peroxidation, and Total Antioxidant Capacity. J. Mol. Neurosci. 2020, 70, 65–70. [Google Scholar] [CrossRef] [PubMed]
  290. 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. [Google Scholar] [CrossRef] [PubMed]
  291. Trojan, E.; Chamera, K.; Bryniarska, N.; Kotarska, K.; Leśkiewicz, M.; Regulska, M.; Basta-Kaim, A. Role of Chronic Administration of Antidepressant Drugs in the Prenatal Stress-Evoked Inflammatory Response in the Brain of Adult Offspring Rats: Involvement of the NLRP3 Inflammasome-Related Pathway. Mol. Neurobiol. 2019, 56, 5365–5380. [Google Scholar] [CrossRef]
  292. Li, J.-M.; Liu, L.-L.; Su, W.-J.; Wang, B.; Zhang, T.; Zhang, Y.; Jiang, C.-L. Ketamine may exert antidepressant effects via suppressing NLRP3 inflammasome to upregulate AMPA receptors. Neuropharmacology 2019, 146, 149–153. [Google Scholar] [CrossRef]
  293. Shao, B.-Z.; Cao, Q.; Liu, C. Targeting NLRP3 Inflammasome in the Treatment of CNS Diseases. Front. Mol. Neurosci. 2018, 11, 320. [Google Scholar] [CrossRef]
  294. Kennedy, S.H.; Lam, R.W.; McIntyre, R.S.; Tourjman, S.V.; Bhat, V.; Blier, P.; Hasnain, M.; Jollant, F.; Levitt, A.J.; MacQueen, G.M.; et al. Canadian Network for Mood and Anxiety Treatments (CANMAT) 2016 Clinical Guidelines for the Management of Adults with Major Depressive Disorder. Can. J. Psychiatry Rev. Can. Psychiatr. 2016, 61, 540–560. [Google Scholar] [CrossRef]
  295. Stokes, P.E. Ten years of fluoxetine. Depress. Anxiety 1998, 8 (Suppl. 1), 1–4. [Google Scholar] [CrossRef]
  296. Stokes, P.E.; Holtz, A. Fluoxetine tenth anniversary update: The progress continues. Clin. Ther. 1997, 19, 1135–1250. [Google Scholar] [CrossRef]
  297. Tranter, R.; O’Donovan, C.; Chandarana, P.; Kennedy, S. Prevalence and outcome of partial remission in depression. J. Psychiatry Neurosci. JPN 2002, 27, 241–247. [Google Scholar]
  298. Mulrow, C.D.; Williams, J.W.; Chiquette, E.; Aguilar, C.; Hitchcock-Noel, P.; Lee, S.; Cornell, J.; Stamm, K. Efficacy of newer medications for treating depression in primary care patients. Am. J. Med. 2000, 108, 54–64. [Google Scholar] [CrossRef] [PubMed]
  299. Kirino, E. Escitalopram for the management of major depressive disorder: A review of its efficacy, safety, and patient acceptability. Patient Prefer. Adherence 2012, 6, 853–861. [Google Scholar] [CrossRef] [PubMed]
  300. Cipriani, A.; Ferla, T.L.; Furukawa, T.A.; Signoretti, A.; Nakagawa, A.; Churchill, R.; McGuire, H.; Barbui, C. Sertraline versus other antidepressive agents for depression. Cochrane Database Syst Rev. 2009, CD006117. [Google Scholar] [CrossRef]
  301. Cipriani, A.; Furukawa, T.A.; Geddes, J.R.; Malvini, L.; Signoretti, A.; McGuire, H.; Churchill, R.; Nakagawa, A.; Barbui, C. MANGA Study Group Does randomized evidence support sertraline as first-line antidepressant for adults with acute major depression? A systematic review and meta-analysis. J. Clin. Psychiatry 2008, 69, 1732–1742. [Google Scholar] [CrossRef]
  302. Gunasekara, N.S.; Noble, S.; Benfield, P. Paroxetine. An update of its pharmacology and therapeutic use in depression and a review of its use in other disorders. Drugs 1998, 55, 85–120. [Google Scholar] [CrossRef]
  303. Sáiz-Ruiz, J.; Ibáñez, A.; Diaz-Marsá, M.; Arias, F.; Padin, J.; Martin-Carrasco, M.; Montes, J.M.; Ferrando, L.; Carrasco, J.L.; Martin-Ballesteros, E.; et al. Efficacy of venlafaxine in major depression resistant to selective serotonin reuptake inhibitors. Prog. Neuropsychopharmacol. Biol. Psychiatry 2002, 26, 1129–1134. [Google Scholar] [CrossRef]
  304. Sanders, B.; Brula, A.Q. Intranasal esketamine: From origins to future implications in treatment-resistant depression. J. Psychiatr. Res. 2021, 137, 29–35. [Google Scholar] [CrossRef]
  305. Chen-Li, D.; Lui, L.M.W.; Rosenblat, J.D.; Lipsitz, O.; Teopiz, K.M.; Ho, R.; Vinberg, M.; Golts, M.; Jawad, M.Y.; Lee, Y.; et al. Ketamine as potential treatment for postpartum depression: A narrative review. Ann. Clin. Psychiatry Off. J. Am. Acad. Clin. Psychiatr. 2022, 34, 264–274. [Google Scholar] [CrossRef]
  306. Włodarczyk, A.; Dywel, A.; Cubała, W.J. Safety and Tolerability of the Acute Ketamine Treatment in Treatment-Resistant Depression: Focus on Comorbidities Interplay with Dissociation and Psychomimetic Symptoms. Pharmaceuticals 2023, 16, 173. [Google Scholar] [CrossRef]
  307. Vasiliu, O. Esketamine for treatment-resistant depression: A review of clinical evidence (Review). Exp. Ther. Med. 2023, 25, 111. [Google Scholar] [CrossRef]
  308. Zhao, L.; Li, Z.; Jin, B.; Hou, N.; Yang, H. Safety and efficacy of low-dose esketamine in laparoscopic cholecystectomy: A prospective, double-blind randomized controlled trial. BMC Anesthesiol. 2024, 24, 47. [Google Scholar] [CrossRef] [PubMed]
  309. Page, M.J.; Moher, D.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E. PRISMA 2020 Explanation and Elaboration: Updated Guidance and Exemplars for Reporting Systematic Reviews. BMJ 2021, 372, n160. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Changes in the main pro- and anti-inflammatory factors in depression. The red arrow represents pro-inflammatory factors and their increase in depression, while the green arrow represents anti-inflammatory factors and their decrease in depression. Created with Biorender.
Figure 1. Changes in the main pro- and anti-inflammatory factors in depression. The red arrow represents pro-inflammatory factors and their increase in depression, while the green arrow represents anti-inflammatory factors and their decrease in depression. Created with Biorender.
Pharmaceuticals 18 00867 g001
Figure 2. Reported effects of SSRIs, SNRIs, ketamine, and esketamine on cytokine levels. Numbers in the columns indicate how many studies found an increase, decrease, or no effect/no significant effect in cytokine levels after treatment with each type of antidepressant. CRP: C-reactive protein; IL-1RA: IL-1 receptor antagonist; TNF-α: Tumor Necrosis Factor Alpha; IFN-γ: interferon gamma.
Figure 2. Reported effects of SSRIs, SNRIs, ketamine, and esketamine on cytokine levels. Numbers in the columns indicate how many studies found an increase, decrease, or no effect/no significant effect in cytokine levels after treatment with each type of antidepressant. CRP: C-reactive protein; IL-1RA: IL-1 receptor antagonist; TNF-α: Tumor Necrosis Factor Alpha; IFN-γ: interferon gamma.
Pharmaceuticals 18 00867 g002
Figure 3. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) [309] flowchart.
Figure 3. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) [309] flowchart.
Pharmaceuticals 18 00867 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bleibel, L.; Sokołowska, P.; Henrykowska, G.; Owczarek, J.; Wiktorowska-Owczarek, A. Unveiling the Anti-Inflammatory Effects of Antidepressants: A Systematic Review of Human Studies over the Last Decade. Pharmaceuticals 2025, 18, 867. https://doi.org/10.3390/ph18060867

AMA Style

Bleibel L, Sokołowska P, Henrykowska G, Owczarek J, Wiktorowska-Owczarek A. Unveiling the Anti-Inflammatory Effects of Antidepressants: A Systematic Review of Human Studies over the Last Decade. Pharmaceuticals. 2025; 18(6):867. https://doi.org/10.3390/ph18060867

Chicago/Turabian Style

Bleibel, Layla, Paulina Sokołowska, Gabriela Henrykowska, Jacek Owczarek, and Anna Wiktorowska-Owczarek. 2025. "Unveiling the Anti-Inflammatory Effects of Antidepressants: A Systematic Review of Human Studies over the Last Decade" Pharmaceuticals 18, no. 6: 867. https://doi.org/10.3390/ph18060867

APA Style

Bleibel, L., Sokołowska, P., Henrykowska, G., Owczarek, J., & Wiktorowska-Owczarek, A. (2025). Unveiling the Anti-Inflammatory Effects of Antidepressants: A Systematic Review of Human Studies over the Last Decade. Pharmaceuticals, 18(6), 867. https://doi.org/10.3390/ph18060867

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop