Next Article in Journal
Leber Hereditary Optic Neuropathy: Molecular Pathophysiology and Updates on Gene Therapy
Next Article in Special Issue
Effect of Voluntary Wheel-Running Exercise on the Endocrine and Inflammatory Response to Social Stress: Conditioned Rewarding Effects of Cocaine
Previous Article in Journal
Identification of Human Breast Adipose Tissue Progenitors Displaying Distinct Differentiation Potentials and Interactions with Cancer Cells
Previous Article in Special Issue
Clinical Value of Inflammatory and Neurotrophic Biomarkers in Bipolar Disorder: A Systematic Review and Meta-Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Relationship between Stress, Inflammation, and Depression

1
Department of Psychiatry, Hanyang University Guri Hospital, Guri 11923, Korea
2
Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
3
Department of Psychiatry, Schulich of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
4
Department of Psychiatry, Hanyang University College of Medicine, Seoul 04763, Korea
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(8), 1929; https://doi.org/10.3390/biomedicines10081929
Submission received: 27 April 2022 / Revised: 29 July 2022 / Accepted: 31 July 2022 / Published: 9 August 2022
(This article belongs to the Special Issue Neuroinflammation in Stress-Related Disorders)

Abstract

:
A narrative review about the relationship between stress, inflammation, and depression is made as follows: Chronic stress leads to various stress-related diseases such as depression. Although most human diseases are related to stress exposure, the common pathways between stress and pathophysiological processes of different disorders are still debatable. Chronic inflammation is a crucial component of chronic diseases, including depression. Both experimental and clinical studies have demonstrated that an increase in the levels of pro-inflammatory cytokines and stress hormones, such as glucocorticoids, substantially contributes to the behavioral alterations associated with depression. Evidence suggests that inflammation plays a key role in the pathology of stress-related diseases; however, this link has not yet been completely explored. In this study, we aimed to determine the role of inflammation in stress-induced diseases and whether a common pathway for depression exists. Recent studies support pharmacological and non-pharmacological treatment approaches significantly associated with ameliorating depression-related inflammation. In addition, major depression can be associated with an activated immune system, whereas antidepressants can exert immunomodulatory effects. Moreover, non-pharmacological treatments for major depression (i.e., exercise) may be mediated by anti-inflammatory actions. This narrative review highlights the mechanisms underlying inflammation and provides new insights into the prevention and treatment of stress-related diseases, particularly depression.

1. Introduction

The pathogenesis of major depression has been mainly explained by the neurogenesis theory for the purpose of overcoming the limitations of the monoamine theory. In terms of the neurogenesis theory, an interaction of serotonergic drugs with postsynaptic 5-HT receptors can be mediated by guanine-nucleotide-binding proteins. In addition, stimulated adenylate cyclase, accompanied by a G-protein, sequentially activates cyclic adenosine monophosphate (cAMP) and increases the level of protein kinase. In contrast, an interaction of noradrenergic drugs with postsynaptic norepinephrine (NE) receptors can inhibit adenylate cyclase. Herein, the second messenger system of 5-HT has a stimulatory effect, whereas that of NE has an inhibitory effect. In addition, increased brain-derived neurotropic factor (BDNF) can alleviate the imbalanced levels of 5-HT and NE in the condition of major depression. However, the other pathogenic process of major depression (i.e., inflammation) or the potential targets of antidepressants have been newly proposed by solving the unmet need for the pharmacological and non-pharmacological treatments of major depression [1,2,3]. Chronic stress can lead to a state of homeostasis dysregulation triggered by environmental and psychological stressors [4,5]. Stressful events activate the sympathetic nervous system and the hypothalamic–pituitary–adrenal (HPA) axis, which engenders multiple neurotransmitters, neurochemicals, and hormonal alterations. The sympathetic nervous system and the HPA axis release chemical mediators that protect the body from stress. Catecholamines increase heart rate and blood pressure, which trigger the “fight or flight” response. In the last two decades, accumulating evidence has indicated that severe stress increases the risk of physical and psychiatric disorders. These are known stress-related diseases. Stress is a common risk factor for over 75% of physical and mental diseases, increasing the morbidity and mortality of these diseases [6]. Psychiatric disorders such as depression are the most common stress-related diseases [6,7,8].
Conventional mechanisms linking stress and disease have focused on the HPA axis and the sympathetic nervous system. However, alterations in the HPA axis and the sympathetic nervous system mainly affect the immune system; thus, the mechanisms linking stress to stress-related diseases remain unclear. Inflammation has emerged as a key mechanism underlying stress [9,10,11,12,13]. Additionally, immune imbalance, especially reflected in reduced natural killer cell activity, has been associated with depressed mood. Furthermore, many studies have indicated that pharmacotherapy has resulted in augmented natural killer cell activity, such as improving depressed mood [14]. A review of the literature suggests that inflammation contributes to the pathophysiology of stress-related disorders. We hypothesized that inflammation is a common factor in stress-related diseases and attempted to elucidate the association between them.

2. Stress and Inflammation

There is evidence that stress can induce inflammatory changes in the brain and peripheral immune system [9,15,16,17,18,19]. Communication between the neuroendocrine and immune systems has been previously reported in different studies [20,21,22,23,24,25]. Stress activates the HPA axis. The hypothalamic secretion of corticotropin-releasing hormones suppresses immune responses by mediating the release of glucocorticoids from the adrenal glands. Studies have revealed that glucocorticoids inhibit lymphocyte cytotoxicity and proliferation [26,27]. Furthermore, they reduce the expression of several pro-inflammatory cytokines, such as interleukin (IL)-6 and tumor necrosis factor (TNF)-α, and they increase the expression of anti-inflammatory cytokines (such as IL-10 and TNF-β) [28,29,30,31,32,33]. However, glucocorticoids also have a pro-inflammatory effect on the immune system [34,35,36,37,38,39]. They enhance the function of the inflammasome NLRP3 through an increase in IL-1β secretion in response to adenosine triphosphate. Inflammasomes are multiple protein complexes that sense external and internal hazardous signals, leading to the cleavage of pro-inflammatory cytokines into mature cytokines, including IL-1β and IL-18. One study revealed that the pro-inflammatory role of glucocorticoids comprises the activation of the innate immune system in response to hazard signals [40,41]. Pro-inflammatory factors, including IL-1, IL-6, and TNF-α, activate the pituitary–adrenal axis, leading to increased serum levels of adrenocorticotropic hormone (ACTH) and glucocorticoids, which, in turn, inhibit the production of pro-inflammatory factors [42,43,44,45,46,47,48]. The immune system and the HPA axis interact to form a negative feedback mechanism. Negative feedback loops can be impaired by a decrease in the expression of cytoplasmic glucocorticoid receptors (GRs) and GR-driven anti-inflammatory genes, resulting in reduced responsiveness to glucocorticoids when stressors overstimulate cytokines [49,50]. In addition to glucocorticoids, the sympathetic nervous system and related neurotransmitters, such as NE and neuropeptide Y (NPY), can affect the immune system and inflammatory function. NE increases the phosphorylation of mitogen-activated protein kinases (MAPKs) through an α-receptor-dependent pathway that activates the secretion of inflammatory factors. Additionally, NPY activates the production of transforming growth factor-β (TGF-β) and TNF-α in macrophage-like cells via the Y1 receptor [51,52,53,54,55].
Both pro- and anti-inflammatory systems are influenced by the intensity and type of stressors [56]. Acute stressors increase immune function, whereas chronic stressors suppress it. Excessive stressors overstimulate the immune system, producing an imbalance between pro- and anti-inflammatory effects. Studies have reported that pro-inflammatory upregulation is induced by stress factors, such as C-reactive protein (CRP), IL-1β, IL-6, TNFα, and nuclear factor kappa B (NF-κB) [57,58,59,60,61,62].
In addition to peripheral inflammation, neuroinflammation has also been reported in response to various stressful conditions [18,63,64,65,66,67,68,69,70]. Pro-inflammatory cytokines that activate microglia lead to the peripheral accumulation of monocytes and macrophages. They have also been found in the brain after stress exposure [71,72,73,74,75,76,77]. Microglia, which are macrophages in the brain, are regarded as the principal source of pro-inflammatory cytokines. Microglia express both glucocorticoid and mineralocorticoid receptors and directly respond to a peak in corticosterone [15,16,78,79]. Additionally, GRs are highly concentrated in the prefrontal cortex and the hippocampus. Thus, stress-related corticosterone indirectly affects microglia. A recent study showed that the innate immune system in the CNS responds to acute stressors by releasing the hazard signal high-mobility group box-1 (HMGB-1) in primate brains, which activates microglia by acting on the NLRP3 inflammasome [80]. Activated microglia exhibit a change in morphology, which is observed as a hypertrophied soma with an enlarged branch. This leads to an increase in cytokines that recruit monocytes from the periphery. An increase in macrophages and peripheral monocytes promotes the production of pro-inflammatory cytokines, such as IL-1β, TNFα, and IL-6, in the brain [81].
Common processes include the activation of the immune system, an increase in sympathetic nervous system pathways, and a decrease in the responsiveness of glucocorticoids, causing the activation of inflammatory responses to stress. Catecholamines, cytokines, glucocorticoids, and other mediators secreted in response to stressors may be the main players in stress-related pro-inflammatory changes.

3. Stress, Inflammation and Depression

Traditionally, inflammation has been considered an essential response to tissue injury or microbial invasion in order to protect homeostasis. Although other pathogeneses are being actively investigated, the molecular basis of the inflammatory pathway is considered critical in the pathogenesis of psychiatric disorders, including depression [82,83,84,85,86]. Increasing evidence suggests that excessive inflammation plays a crucial role in the onset and progression of stress-related diseases. Evidence supports the idea that the inflammatory response forms the background of multifactorial diseases, including psychiatric disorders [13,87,88,89,90].
Stressful events are elemental in inciting episodes of major depressive disorder (MDD). Patients with depression are predisposed to the activation of the HPA axis and hypercortisolemia, rising levels of stress hormones and corticotropin-releasing hormone, and an increase in ACTH [91,92,93,94]. MAPKs enhance the activity of serotonin membrane transporters, and serotonin is the most vital neurotransmitter related to depression [95,96,97,98].
Various concepts of inflammation, such as the macrophage hypothesis and the cytokine theory, have been suggested as the underlying pathology in MDD. The main principle of inflammatory depression is the activation of the immune response, particularly cytokine production. This affects the levels of neurochemicals that lead to MDD [99]. Stress can promote the development of depression-like behavior by facilitating the expression of inflammatory cytokines [100,101]. Recently, the novel kynurenine pathway (KP) has gained attention in the cytokine theory. Pro-inflammatory cytokines activate KP to influence tryptophan metabolism and secrete neurotoxins, which may either decrease serotonin production or promote serotonin reuptake [102,103].
Studies on patients and animal models have supported inflammation as a pathology of depression. Inflammatory cytokines, such as IL-1β, TNF-α, and IFN-α, or inducers, such as vaccinations, have been found to result in alterations in human and rodent behaviors. Increased levels of inflammatory mediators, including cytokines, acute phase proteins, chemokines, adhesion molecules, and prostaglandins, have also been found in the CNS, blood, and cerebrospinal fluid of patients with depression [57,100,104,105,106,107]. Chronic stress is frequently used to establish depression models. Exposure to chronic stress for four weeks significantly activates inflammatory cytokines, including IL-18, IL-1β, TNF-α, and inflammatory inducible nitric oxide synthase (NOS) [108]. With the upregulation of pro-inflammatory cytokines, depression-like behaviors are provoked. Blocking inflammatory cytokines or inducible NOS with agents such as minocycline helps eliminate the depression-like behavior caused by stress [108]. Several antidepressants have been shown to have anti-inflammatory effects. Antidepressants and nonsteroidal anti-inflammatory drugs, such as minocycline, mitigate microglial activation and reduce blood levels of IL-6 and central cytokine secretion, thereby diminishing behavioral alterations [109].
Inflammasomes are multiple protein complexes that drive the production and maturation of pro-inflammatory factors, such as IL-18 and IL-1β, to induce innate immune defenses [110,111]. The NLRP3 inflammasome has been implicated in the induction of depression-like behaviors in a lipopolysaccharide (LPS)-induced mouse model [112,113]. A recent study demonstrated that the protective role of caspase-1 inhibition in brain function and gut microbiota provoked depression-like and anxiety-like behaviors [114,115,116,117].

4. MDD Is Associated with an Activated Immune System

MDD is associated with diseases characterized by an activated immune system, such as autoimmune diseases (systemic lupus erythematosus (SLE), type 1 diabetes, and rheumatoid arthritis (RA)), allergies, and infections (sepsis). Patients with both asthma and atopy have an approximately 50% higher incidence of MDD [115,116]. Approximately, 36% of patients with asthma suffer from MDD, and their TNFα levels are significantly increased, while their IFNγ levels are significantly decreased.
Meta-analyses have revealed that the incidence of MDD in patients with diabetes is up to twice that in those without diabetes [118,119]. Inflammatory activation is linked to the pathogenesis of diabetes, with the immune response implicated in both type 1 and type 2 diabetes [120]. Inflammatory markers, including CRP, IL-1β, IL-1RA, and MCP-1, are significantly increased in patients with MDD and type 2 diabetes [121].
One-third of the patients with SLE were reported to suffer from MDD in another meta-analysis that used the standard depression scale subscale for depression and hospital anxiety [122]. Studies have also shown that severe fatigue is associated with a high risk of MDD and that there is no correlation with SLE severity [123,124]. A review found that over 90% of patients with SLE experience fatigue without an association with the severity of SLE [125]. Increased TNF-α and decreased IL-10 levels have been demonstrated in patients with SLE and MDD and have been associated with a more severe course of MDD [126,127].
MDD is also more prevalent in patients with RA. Studies have reported a 74% higher risk of RA in patients with MDD than in healthy controls. In addition, 17% of patients with RA have MDD [128,129]. More than 70% of patients with RA experience clinically significant fatigue [130]. A study reported a positive association between serum CRP levels and MDD severity among patients with RA [131]. CRP levels and the erythrocyte sedimentation rate, a marker of inflammation severity, are significantly associated with clinically significant fatigue [132]. A review investigated diverse anti-TNF and other biological agents in patients with RA and reported that they had significant impacts on fatigue felt by patients, further demonstrating that fatigue may partly underpin immune responses [133].
The association between immune activation and MDD has not only been reported in immune-related disorders but also in cases of infections. Sepsis leads to broad pro-inflammatory reactions that trigger a systemic immune response to an infective agent. Patients with sepsis have a substantially increased level of inflammatory markers, even after sepsis resolution [134,135,136]. Sepsis survivors have an increased rate of MDD compared to normal controls; however, this has been found to not be significantly higher than the rates of MDD preceding infection [137]. This high rate of MDD in patients with sepsis also reflects that psychological stress increases MDD and immune activation [138] and is related to a higher risk of sepsis [139]. Few studies have reported post-sepsis MDD in human patients, while animal studies have reported sepsis conditions leading to changes in emotion [140]. Animal studies have also reported that immune suppression, by suppressing the NF-κB pathway, reduces MDD-like behaviors in animal models [140,141]. There may be a possible role for the “priming” of the immune response by conditions such as sepsis, in turn triggering a higher risk of developing MDD [142]. Future studies are warranted to elucidate whether immune activation predisposes the immune system to be more reactive to stress and insults, resulting in an increased risk of MDD.
Recently, a bioinformatics analytical approach has gained prevalence using peripheral blood biomarkers by screening for potential mRNAs in patients with MDD [143]. Zhang et al.,used public data from an mRNA microarray to screen for hub gene alterations implicated in MDD. Differentially expressed genes were analyzed using the Kyoto Encyclopedia of Genes and Genomes and Gene Ontology pathway datasets. A combined gene set of co-upregulated differentially expressed genes was associated with immune response, neutrophil activation, degranulation, cell-mediated immunity, and the Toll-like receptor signaling pathway.

5. Antidepressants Exert Immunomodulatory Effects

The response to antidepressants has been associated with changes in immune marker levels. In animal models treated with LPS, low serum levels of TNFα and high levels of IL-10 were identified following the administration of an serotonin reuptake inhibitor (SSRI) and a serotonin–norepinephrine reuptake inhibitor (SNRI) [144]. In a social stress model, tricyclic antidepressant (TCA) administration reduced microglial IL-6 mRNA in vivo and ex vivo and decreased TNF-α and IL-1β mRNA levels [145]. Studies using macrophages in animal models have also found a similar immunosuppressive influence, in which the reduction in IL-6 and enhancement in IL-10 levels following antidepressant administration indicated that such impacts might be mediated by the inhibition of the NF-κB pathway [146]. Meanwhile, a study revealed that the administration of SSRIs and mirtazapine resulted in the opposite effect on cytokine production, with increased levels of inflammatory markers, including IL-6, IL-1β, and TNF-α [147]. Human studies investigating alterations in cytokine levels have found that the administration of antidepressant agents decreased the levels of IL-1β, IL-4, IL-6, and IL-10 [148,149]. Other studies have shown that antidepressants have different immunomodulatory effects. SNRIs, such as venlafaxine, have a higher anti-inflammatory effect than SSRIs [150]. This study also demonstrated that SSRI administration significantly enhanced serum IL-6 and TNFα levels. Other studies have also found that psychotherapy has an immunomodulatory effect similar to that of pharmaceutical treatment [151]. Recent studies focused on physical exercise and transcranial direct current stimulation have reported that circulating cytokine levels decreased following treatment. However, there are no consistent findings regarding the improvement in MDD symptoms [152,153]. The administration of electroconvulsive therapy (ECT) has a similar effect on the immune system. ECT is linked to an initial increase in IL-1 and IL-6, with the levels of TNFα and IL-6 decreasing after long-term treatment. However, these results need to be further replicated [154]. One study focused on the effect of ECT as an adjunctive to antidepressant agents and reported that, while it significantly reduced IL-6 levels, TNFα levels increased after treatment [155]. ECT has also been reported to reverse changes in NK cell activity, which is decreased in patients with MDD [156].
Studies have also demonstrated that immune markers can be used to predict therapeutic efficacy. Low levels of pro-inflammatory cytokines can predict better therapeutic responses to SSRIs, TCAs, and ketamine, and responders may have significantly reduced cytokine levels [157,158]. However, serum CRP levels predict different therapeutic responses to different antidepressant agents [159]. Patients with reduced CRP levels showed a better response to escitalopram, while those with increased CRP levels demonstrated a better response to TCA, such as nortriptyline. These findings suggest that SSRI effects may be partly derived from anti-inflammatory actions. High IL-6 levels in patients have also been correlated with worse therapeutic efficacy of different SSRIs and SNRIs [160]. However, higher TNF-α levels were correlated with therapeutic non-response in patients treated with escitalopram [161].
In sleep deprivation therapy, an antidepressant treatment option, high IL-6 levels predict worse therapeutic responses in patients with MDD [162]. Reduced TNF-α levels at the first ECT have also been shown to predict therapeutic outcomes [163]. However, the link between high levels of inflammatory cytokines and poor therapeutic efficacy has not been confirmed for all therapeutic options. It has been demonstrated that high pro-inflammatory cytokine levels, such as TNFα, predict a favorable response to physical exercise [153]. The differences in the predictive effects of inflammatory cytokine levels in light of the different therapeutic efficacies indicate that their underlying mechanisms may differ, with anti-inflammatory influences being more crucial for some therapeutic options, such as SSRIs, than others.
Fatigue may be effectively controlled with a few medications. However, few studies have investigated the association between these medications and the immune system. Amantadine can be effective in patients with multiple sclerosis [164]; however, little research has focused on its immunomodulatory effects. Amantadine administration in rats enhanced the effect of fluoxetine, an SSRI, when co-administered; however, it did not alter IFNγ or IL-10 levels [165]. Further studies are warranted to examine whether its therapeutic efficacy for fatigue in patients may act via its effects on the immune system.

6. Non-Pharmacological Treatment Effects on Inflammatory Depression

Omega-3 fatty acids have been identified as potential treatments for MDD-related inflammations. Patients with MDD have low levels of n-3 polyunsaturated fatty acids (PUFA) [166], indicating that n-3 PUFAs may be involved in MDD pathophysiology. Most randomized controlled trials investigating the antidepressant effects of n-3 PUFAs have shown small-to-medium effects [165,166,167,168,169], as only patients with signs of low-grade inflammation responded to this PUFA intervention. This may be consistent with a study showing that n-3 PUFA administration hinders LPS-induced depression-like behavior in mice via the suppression of neuroinflammation [170]. Another study reported that n-3 PUFA administration had a protective effect on IFN-α-induced MDD, further supporting the role of n-3 PUFA in MDD-related inflammation [171]. Exercise therapy may be an appropriate alternative to conventional antidepressants [172,173,174]. Interestingly, recent studies have shown that the antidepressant effects of physical exercise may be mediated by anti-inflammatory actions [175]. A clinical study demonstrated that patients with MDD who partially responded to SSRIs showed a significant decrease in TNF-α levels after 12 weeks of physical exercise [153].

7. Limitations and Future Directions

The literature regarding stress-related inflammation reviewed here may be useful for understanding the common physiology of stress-related diseases. However, there are unanswered questions that require further investigation. In addition to inflammation, crosstalk between inflammation and other related pathways, such as cell stress, has not been extensively investigated. Moreover, there have been very few studies on the correlation between specific pathways and stress-related diseases. It is unknown whether other pathways or mechanisms that do not modify the peripheral immune system can affect neuroinflammation in depression. From a clinical perspective, anti-inflammatory treatment must target specific cells and pathways in the brain that are crucial in the pathogenesis of depression. Further research is required to address these limitations, which will contribute to the development of novel therapies for stress-related diseases, particularly depression. To improve stress, psychological and physical stressors related to multifactorial mechanisms should be prevented in patients with depression. Moreover, targeting stress-related risk factors comprising stress-induced inflammation could be beneficial in preventing diseases among highly stressful individuals, such as those with depression.

8. Conclusions

Stress prompts inflammation not only peripherally but also centrally through the dysregulation of the immune system (Figure 1). This results in the development of stress-related diseases, particularly depression. Although there are various facilitating factors, inflammation appears to be the central pathology that leads to depression. In this review, we provide evidence that stress induces depression through neuroinflammation and peripheral inflammation. Stress weakens the central microglia, blood, and immune system by activating the sympathetic nervous system and the HPA axis. Thus, our findings support the notion that inflammation is the main pathway for stress-related diseases, such as depression, and that it contributes to disease progression by affecting the early course of the disease. The altered activity of the hypothalamic–pituitary–adrenal (HPA) axis, the weakened blood–brain barrier (BBB), and the facilitated passage of pro-inflammatory cytokines may be related to the serotonergic, noradrenergic, and dopaminergic dysfunction of major depression [176,177,178]. In addition, the tryptophan–kynurenine metabolic pathway is involved in the process of chronic neuroinflammation, which is associated with major depression, in the context of pro-inflammatory cytokines and the immune response [179,180]. Moreover, PUFAs have the same neuroprotective effects as non-pharmacological treatments for depression, based on their effects on microglial polarization [181].

Author Contributions

Conceptualization, I.-B.K. and S.-C.P.; methodology, I.-B.K.; investigation, I.-B.K.; resources, I.-B.K.; writing—original draft preparation, I.-B.K. and S.-C.P.; writing—review and editing, I.-B.K., J.-H.L. and S.-C.P.; visualization, I.-B.K., J.-H.L. and S.-C.P.; supervision, J.-H.L. and S.-C.P.; project administration, J.-H.L. and S.-C.P. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the research fund of Hanyang University (HY-202100000003685).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Shen, W.W. Clinical Psychopharmacology for the 21st Century, 3rd ed.; Ho-Chi Publishing Company: Taipei, Taiwan, 2011. [Google Scholar]
  2. Drazinix, C.M.; Sazbo, S.T. Neurotramitters and Receptors in Psychiatric Disorder (Chapter 2). In The American Psychiatric Association Publishing Textbook of Psychopharmacology; Schatzberg, A.F., Nemeroff, C.B., Eds.; APA Publishing: Washington, DC, USA, 2017. [Google Scholar]
  3. Jessberger, S.; Kempermann, G. Adult-born hippocampal neurons mature into activity-dependent responsiveness. Eur. J. Neurosci. 2003, 18, 2707–2712. [Google Scholar] [CrossRef]
  4. Landsbergis, P.A. The changing organization of work and the safety and health of working people: A commentary. J. Occup. Environ Med. 2003, 45, 61–72. [Google Scholar] [CrossRef]
  5. Molina, P.E. Rethinking Integration of Environmental and Behavioral Stressors; Back to Energy Homeostasis and Function. Function 2022, 3, zqab074. [Google Scholar] [CrossRef]
  6. Cohen, S.; Janicki-Deverts, D.; Miller, G.E. Psychological stress and disease. JAMA 2007, 298, 1685–1687. [Google Scholar] [CrossRef]
  7. Cohen, S.; Gianaros, P.J.; Manuck, S.B. A stage model of stress and disease. Perspect. Psychol. Sci. 2016, 11, 456–463. [Google Scholar] [CrossRef]
  8. Turner, A.I.; Smyth, N.; Hall, S.J.; Torres, S.J.; Hussein, M.; Jayasinghe, S.U.; Ball, K.; Clow, A.J. Psychological stress reactivity and future health and disease outcomes: A systematic review of prospective evidence. Psychoneuroendocrinology 2020, 114, 104599. [Google Scholar] [CrossRef]
  9. Rohleder, N. Stimulation of systemic low-grade inflammation by psychosocial stress. Psychosom. Med. 2014, 76, 181–189. [Google Scholar] [CrossRef]
  10. Rohleder, N. Stress and inflammation—The need to address the gap in the transition between acute and chronic stress effects. Psychoneuroendocrinology 2019, 105, 164–171. [Google Scholar] [CrossRef]
  11. Viljoen, M.; Lee Thomas neé Negrao, B. Low-grade systemic inflammation and the workplace. Work 2021, 69, 903–915. [Google Scholar] [CrossRef]
  12. Hantsoo, L.; Kornfield, S.; Anguera, M.C.; Epperson, C.N. Inflammation: A proposed intermediary between maternal stress and offspring neuropsychiatric risk. Biol. Psychiatr. 2019, 85, 97–106. [Google Scholar] [CrossRef]
  13. Furman, D.; Campisi, J.; Verdin, E.; Carrera-Bastos, P.; Targ, S.; Franceschi, C.; Ferrucci, L.; Gilroy, D.W.; Fasano, A.; Miller, G.W. Chronic inflammation in the etiology of disease across the life span. Nat. Med. 2019, 25, 1822–1832. [Google Scholar] [CrossRef] [PubMed]
  14. Frank, M.G.; Hendricks, S.E.; Johnson, D.R.; Wieseler, J.; Burke, W.J. Antidepressants augment natural killer cell activity: In vivo and in vitro. Neuropsychobiology 1999, 39, 18–24. [Google Scholar] [CrossRef] [PubMed]
  15. Calcia, M.A.; Bonsall, D.R.; Bloomfield, P.S.; Selvaraj, S.; Barichello, T.; Howes, O.D. Stress and neuroinflammation: A systematic review of the effects of stress on microglia and the implications for mental illness. Psychopharmacology 2016, 233, 1637–1650. [Google Scholar] [CrossRef] [Green Version]
  16. Picca, A.; Calvani, R.; Coelho-Junior, H.J.; Landi, F.; Bernabei, R.; Marzetti, E. Mitochondrial dysfunction, oxidative stress, and neuroinflammation: Intertwined roads to neurodegeneration. Antioxidants 2020, 9, 647. [Google Scholar] [CrossRef]
  17. Black, P.H. Stress and the inflammatory response: A review of neurogenic inflammation. Brain Behav. Immun. 2002, 16, 622–653. [Google Scholar] [CrossRef]
  18. Liu, Y.-Z.; Wang, Y.-X.; Jiang, C.-L. Inflammation: The common pathway of stress-related diseases. Front. Hum. Neurosci. 2017, 11, 316. [Google Scholar] [CrossRef]
  19. Liezmann, C.; Klapp, B.; Peters, E. Stress, atopy and allergy: A re-evaluation from a psychoneuroimmunologic persepective. Dermato Endocrinology 2011, 3, 37–40. [Google Scholar] [CrossRef] [Green Version]
  20. Jiang, C.-L.; Lu, C.-L.; Liu, X.-Y. The molecular basis for bidirectional communication between the immune and neuroendocrine systems. Domest. Anim. Endocrinol. 1998, 15, 363–369. [Google Scholar] [CrossRef]
  21. Quan, N.; Banks, W.A. Brain-immune communication pathways. Brain Behav. Immun. 2007, 21, 727–735. [Google Scholar] [CrossRef]
  22. Arambula, S.E.; McCarthy, M.M. Neuroendocrine-immune crosstalk shapes sex-specific brain development. Endocrinology 2020, 161, bqaa055. [Google Scholar] [CrossRef] [Green Version]
  23. Quatrini, L.; Vivier, E.; Ugolini, S. Neuroendocrine regulation of innate lymphoid cells. Immunol. Rev. 2018, 286, 120–136. [Google Scholar] [CrossRef] [Green Version]
  24. Weigent, D.A.; Blalock, J.E. Associations between the neuroendocrine and immune systems. J. Leukoc. Biol. 1995, 58, 137–150. [Google Scholar] [CrossRef]
  25. Weigent, D.A.; Carr, D.; Blalock, J.E. Bidirectional communication between the neuroendocrine and immune systems. Common hormones and hormone receptors. Ann. N. Y. Acad. Sci. 1990, 579, 17–27. [Google Scholar] [CrossRef] [PubMed]
  26. Gallelli, L.; D’Agostino, B.; Vatrella, A.; Fratto, D.; Renda, T.; Galderisi, U.; Piegari, E.; Crimi, N.; Rossi, F.; Caputi, M. Effects of TGF-beta and glucocorticoids on map kinase phosphorylation, IL-6/IL-11 secretion and cell proliferation in primary cultures of human lung fibroblasts. J. Cell. Physiol. 2007, 210, 489–497. [Google Scholar]
  27. Gallelli, L.; Pelaia, G.; Fratto, D.; Muto, V.; Falcone, D.; Vatrella, A.; Curto, L.; Renda, T.; Busceti, M.; Liberto, M. Effects of budesonide on p38 MAPK activation, apoptosis and IL-8 secretion, induced by TNF-α and Haemophilus influenzae in human bronchial epithelial cells. Int. J. Immunopathol. Pharmacol. 2010, 23, 471–479. [Google Scholar] [CrossRef]
  28. Sorrells, S.F.; Caso, J.R.; Munhoz, C.D.; Sapolsky, R.M. The stressed CNS: When glucocorticoids aggravate inflammation. Neuron 2009, 64, 33–39. [Google Scholar] [CrossRef] [Green Version]
  29. Hill, A.R.; Spencer-Segal, J.L. Glucocorticoids and the brain after critical illness. Endocrinology 2021, 162, bqaa242. [Google Scholar] [CrossRef]
  30. Liberman, A.C.; Trias, E.; da Silva Chagas, L.; Trindade, P.; dos Santos Pereira, M.; Refojo, D.; Hedin-Pereira, C.; Serfaty, C.A. Neuroimmune and inflammatory signals in complex disorders of the central nervous system. Neuroimmunomodulation 2018, 25, 246–270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Besedovsky, H.O.; Del Rey, A. Regulating inflammation by glucocorticoids. Nat. Immunol. 2006, 7, 537. [Google Scholar] [CrossRef]
  32. Duque, E.D.A.; Munhoz, C.D. The pro-inflammatory effects of glucocorticoids in the brain. Front. Endocrinol. 2016, 7, 78. [Google Scholar] [CrossRef] [Green Version]
  33. Sorrells, S.F.; Munhoz, C.D.; Manley, N.C.; Yen, S.; Sapolsky, R.M. Glucocorticoids increase excitotoxic injury and inflammation in the hippocampus of adult male rats. Neuroendocrinology 2014, 100, 129–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Elenkov, I.J. Neurohormonal-cytokine interactions: Implications for inflammation, common human diseases and well-being. Neurochem. Int. 2008, 52, 40–51. [Google Scholar] [CrossRef] [PubMed]
  35. van den Heuvel, L.L.; Suliman, S.; Bröcker, E.; Kilian, S.; Stalder, T.; Kirschbaum, C.; Seedat, S. The association between hair cortisol levels, inflammation and cognitive functioning in females. Psychoneuroendocrinology 2022, 136, 105619. [Google Scholar] [CrossRef]
  36. Cruz-Topete, D.; Cidlowski, J.A. One hormone, two actions: Anti-and pro-inflammatory effects of glucocorticoids. Neuroimmunomodulation 2015, 22, 20–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Oppong, E.; Cato, A.C. Effects of glucocorticoids in the immune system. Adv. Exp. Med. Biol. 2015, 872, 217–233. [Google Scholar]
  38. Frank, M.G.; Thompson, B.M.; Watkins, L.R.; Maier, S.F. Glucocorticoids mediate stress-induced priming of microglial pro-inflammatory responses. Brain Behav. Immun. 2012, 26, 337–345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Cain, D.W.; Cidlowski, J.A. Immune regulation by glucocorticoids. Nat. Rev. Immunol. 2017, 17, 233–247. [Google Scholar] [CrossRef]
  40. Busillo, J.M.; Azzam, K.M.; Cidlowski, J.A. Glucocorticoids sensitize the innate immune system through regulation of the NLRP3 inflammasome. J. Biol. Chem. 2011, 286, 38703–38713. [Google Scholar] [CrossRef] [Green Version]
  41. Gülke, E.; Gelderblom, M.; Magnus, T. Danger signals in stroke and their role on microglia activation after ischemia. Ther. Adv. Neurol. Disord. 2018, 11, 1756286418774254. [Google Scholar] [CrossRef] [Green Version]
  42. Alley, D.E.; Seeman, T.E.; Kim, J.K.; Karlamangla, A.; Hu, P.; Crimmins, E.M. Socioeconomic status and C-reactive protein levels in the US population: NHANES IV. Brain Behav. Immun. 2006, 20, 498–504. [Google Scholar] [CrossRef]
  43. Danese, A.; Pariante, C.M.; Caspi, A.; Taylor, A.; Poulton, R. Childhood maltreatment predicts adult inflammation in a life-course study. Proc. Natl. Acad. Sci. USA 2007, 104, 1319–1324. [Google Scholar] [CrossRef] [Green Version]
  44. Steptoe, A.; Hamer, M.; Chida, Y. The effects of acute psychological stress on circulating inflammatory factors in humans: A review and meta-analysis. Brain Behav. Immun. 2007, 21, 901–912. [Google Scholar] [CrossRef] [PubMed]
  45. Miller, G.E.; Chen, E.; Sze, J.; Marin, T.; Arevalo, J.M.; Doll, R.; Ma, R.; Cole, S.W. A functional genomic fingerprint of chronic stress in humans: Blunted glucocorticoid and increased NF-κB signaling. Biol. Psychiatr. 2008, 64, 266–272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Elenkov, I.J.; Chrousos, G.P. Stress system–organization, physiology and immunoregulation. Neuroimmunomodulation 2006, 13, 257–267. [Google Scholar] [CrossRef]
  47. Papargyri, P.; Zapanti, E.; Salakos, N.; Papargyris, L.; Bargiota, A.; Mastorakos, G. Links between HPA axis and adipokines: Clinical implications in paradigms of stress-related disorders. Expert Rev. Endocrinol. Metab. 2018, 13, 317–332. [Google Scholar] [CrossRef]
  48. Borghetti, P.; Saleri, R.; Mocchegiani, E.; Corradi, A.; Martelli, P. Infection, immunity and the neuroendocrine response. Vet. Immunol. Immunopathol. 2009, 130, 141–162. [Google Scholar] [CrossRef] [PubMed]
  49. Sterling, P. Allostasis: A new paradigm to explain arousal pathology. In Handbook of Life Stress, Cognition and Health; John Wiley & Sons: New York, NY, USA, 1988; pp. 629–649. [Google Scholar]
  50. Lee, S.W. A Copernican approach to brain advancement: The paradigm of allostatic orchestration. Front. Hum. Neurosci. 2019, 13, 129. [Google Scholar] [CrossRef]
  51. Bellinger, D.L.; Millar, B.A.; Perez, S.; Carter, J.; Wood, C.; ThyagaRajan, S.; Molinaro, C.; Lubahn, C.; Lorton, D. Sympathetic modulation of immunity: Relevance to disease. Cell Immunol. 2008, 252, 27–56. [Google Scholar] [CrossRef] [Green Version]
  52. Zhou, J.-R.; Xu, Z.; Jiang, C.-L. Neuropeptide Y promotes TGF-β1 production in RAW264. 7 cells by activating PI3K pathway via Y1 receptor. Neurosci. Bull. 2008, 24, 155–159. [Google Scholar] [CrossRef]
  53. Huang, J.-L.; Zhang, Y.-L.; Wang, C.-C.; Zhou, J.-R.; Ma, Q.; Wang, X.; Shen, X.-H.; Jiang, C.-L. Enhanced phosphorylation of MAPKs by NE promotes TNF-α production by macrophage through α adrenergic receptor. Inflammation 2012, 35, 527–534. [Google Scholar] [CrossRef]
  54. Bellinger, D.L.; Lorton, D. Sympathetic nerve hyperactivity in the spleen: Causal for nonpathogenic-driven chronic immune-mediated inflammatory diseases (IMIDs)? Int. J. Mol. Sci. 2018, 19, 1188. [Google Scholar] [CrossRef] [Green Version]
  55. Brinkman, D.J.; Ten Hove, A.S.; Vervoordeldonk, M.J.; Luyer, M.D.; de Jonge, W.J. Neuroimmune interactions in the gut and their significance for intestinal immunity. Cells 2019, 8, 670. [Google Scholar] [CrossRef] [Green Version]
  56. Song, C.; Kenis, G.; van Gastel, A.; Bosmans, E.; Lin, A.; de Jong, R.; Neels, H.; Scharpé, S.; Janca, A.; Yasukawa, K. Influence of psychological stress on immune-inflammatory variables in normal humans. Part II. Altered serum concentrations of natural anti-inflammatory agents and soluble membrane antigens of monocytes and T lymphocytes. Psychiatr. Res. 1999, 85, 293–303. [Google Scholar] [CrossRef]
  57. Miller, A.H.; Maletic, V.; Raison, C.L. Inflammation and its discontents: The role of cytokines in the pathophysiology of major depression. Biol. Psychiatr. 2009, 65, 732–741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. 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] [PubMed]
  59. Draganov, M.; Arranz, M.J.; Salazar, J.; de Diego-Adeliño, J.; Gallego-Fabrega, C.; Jubero, M.; Carceller-Sindreu, M.; Portella, M.J. Association study of polymorphisms within inflammatory genes and methylation status in treatment response in major depression. Eur. Psychiatr. 2019, 60, 7–13. [Google Scholar] [CrossRef] [PubMed]
  60. Opel, N.; Cearns, M.; Clark, S.; Toben, C.; Grotegerd, D.; Heindel, W.; Kugel, H.; Teuber, A.; Minnerup, H.; Berger, K. Large-scale evidence for an association between low-grade peripheral inflammation and brain structural alterations in major depression in the BiDirect study. J. Psychiatr. Neurosci. 2019, 44, 423–431. [Google Scholar] [CrossRef] [Green Version]
  61. Halaris, A. Inflammation and depression but where does the inflammation come from? Curr. Opin. Psychiatr. 2019, 32, 422–428. [Google Scholar] [CrossRef]
  62. Rhie, S.J.; Jung, E.-Y.; Shim, I. The role of neuroinflammation on pathogenesis of affective disorders. J. Exerc. Rehabil. 2020, 16, 2. [Google Scholar] [CrossRef] [Green Version]
  63. García-Bueno, B.; Caso, J.R.; Leza, J.C. Stress as a neuroinflammatory condition in brain: Damaging and protective mechanisms. Neurosci. Biobehav. Rev. 2008, 32, 1136–1151. [Google Scholar] [CrossRef]
  64. Gu, M.; Mei, X.-L.; Zhao, Y.-N. Sepsis and cerebral dysfunction: BBB damage, neuroinflammation, oxidative stress, apoptosis and autophagy as key mediators and the potential therapeutic approaches. Neurotox. Res. 2021, 39, 489–503. [Google Scholar] [CrossRef]
  65. He, J.; Zhu, G.; Wang, G.; Zhang, F. Oxidative stress and neuroinflammation potentiate each other to promote progression of dopamine neurodegeneration. Oxid. Med. Cell Longev. 2020, 6137521. [Google Scholar] [CrossRef] [PubMed]
  66. Solleiro-Villavicencio, H.; Rivas-Arancibia, S. Effect of chronic oxidative stress on neuroinflammatory response mediated by CD4+ T cells in neurodegenerative diseases. Front. Cell Neurosci. 2018, 12, 114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Munhoz, C.; Garcia-Bueno, B.; Madrigal, J.; Lepsch, L.; Scavone, C.; Leza, J. Stress-induced neuroinflammation: Mechanisms and new pharmacological targets. Braz. J. Med. Biol. Res. 2008, 41, 1037–1046. [Google Scholar] [CrossRef]
  68. Gárate, I.; Garcia-Bueno, B.; Madrigal, J.L.M.; Caso, J.R.; Alou, L.; Gomez-Lus, M.L.; Micó, J.A.; Leza, J.C. Stress-induced neuroinflammation: Role of the Toll-like receptor-4 pathway. Biol. Psychiatr. 2013, 73, 32–43. [Google Scholar] [CrossRef] [PubMed]
  69. Taylor, J.M.; Main, B.S.; Crack, P.J. Neuroinflammation and oxidative stress: Co-conspirators in the pathology of Parkinson’s disease. Neurochem. Int. 2013, 62, 803–819. [Google Scholar] [CrossRef] [PubMed]
  70. Hori, H.; Kim, Y. Inflammation and post-traumatic stress disorder. Psychiatr. Clin. Neurosci. 2019, 73, 143–153. [Google Scholar] [CrossRef] [Green Version]
  71. Johnson, J.; Campisi, J.; Sharkey, C.; Kennedy, S.; Nickerson, M.; Greenwood, B.; Fleshner, M. Catecholamines mediate stress-induced increases in peripheral and central inflammatory cytokines. Neuroscience 2005, 135, 1295–1307. [Google Scholar] [CrossRef]
  72. Johnson, J.D.; Barnard, D.F.; Kulp, A.C.; Mehta, D.M. Neuroendocrine regulation of brain cytokines after psychological stress. J. Endocr. Soc. 2019, 3, 1302–1320. [Google Scholar] [CrossRef]
  73. Barnard, D.F. The Regulation of Brain Pro-Inflammatory Cytokines: Implications for Stress and Depression. Ph.D. Thesis, Kent State University, Kent, OH, USA, 2020. [Google Scholar]
  74. Foertsch, S.; Reber, S.O. The role of physical trauma in social stress-induced immune activation. Neurosci. Biobehav. Rev. 2020, 113, 169–178. [Google Scholar] [CrossRef] [PubMed]
  75. Wohleb, E.S.; Franklin, T.; Iwata, M.; Duman, R.S. Integrating neuroimmune systems in the neurobiology of depression. Nat. Rev. Neurosci. 2016, 17, 497–511. [Google Scholar] [CrossRef] [PubMed]
  76. Weber, M.D.; Godbout, J.P.; Sheridan, J.F. Repeated social defeat, neuroinflammation, and behavior: Monocytes carry the signal. Neuropsychopharmacology 2017, 42, 46–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Ramirez, K.; Fornaguera-Trías, J.; Sheridan, J.F. Stress-induced microglia activation and monocyte trafficking to the brain underlie the development of anxiety and depression. Inflamm. Assoc. Depress. Evid. Mech. Implic. 2016, 31, 155–172. [Google Scholar]
  78. Mendiola, A.S.; Ryu, J.K.; Bardehle, S.; Meyer-Franke, A.; Ang, K.K.-H.; Wilson, C.; Baeten, K.M.; Hanspers, K.; Merlini, M.; Thomas, S. Transcriptional profiling and therapeutic targeting of oxidative stress in neuroinflammation. Nat. Immunol. 2020, 21, 513–524. [Google Scholar] [CrossRef] [PubMed]
  79. Fonken, L.K.; Weber, M.D.; Daut, R.A.; Kitt, M.M.; Frank, M.G.; Watkins, L.R.; Maier, S.F. Stress-induced neuroinflammatory priming is time of day dependent. Psychoneuroendocrinology 2016, 66, 82–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Weber, M.D.; Frank, M.G.; Tracey, K.J.; Watkins, L.R.; Maier, S.F. Stress induces the danger-associated molecular pattern HMGB-1 in the hippocampus of male Sprague Dawley rats: A priming stimulus of microglia and the NLRP3 inflammasome. J. Neurosci. 2015, 35, 316–324. [Google Scholar] [CrossRef]
  81. Wohleb, E.S.; Delpech, J.-C. Dynamic cross-talk between microglia and peripheral monocytes underlies stress-induced neuroinflammation and behavioral consequences. Prog. Neuropsychopharmacol. Biol. Psychiatr. 2017, 79, 40–48. [Google Scholar] [CrossRef]
  82. Park, S.-C. Neurogenesis and antidepressant action. Cell Tissue Res. 2019, 377, 95–106. [Google Scholar] [CrossRef]
  83. Kim, Y.-K.; Park, S.-C. An alternative approach to future diagnostic standards for major depressive disorder. Prog. Neuropsychopharmacol. Biol. Psychiatr. 2021, 105, 110133. [Google Scholar] [CrossRef]
  84. Kim, I.B.; Park, S.-C. Neural Circuitry–Neurogenesis Coupling Model of Depression. Int. J. Mol. Sci. 2021, 22, 2468. [Google Scholar] [CrossRef]
  85. Kim, I.B.; Park, S.-C. Machine Learning-Based Definition of Symptom Clusters and Selection of Antidepressants for Depressive Syndrome. Diagnostics 2021, 11, 1631. [Google Scholar] [CrossRef] [PubMed]
  86. Kim, I.B.; Park, S.-C. The Entorhinal Cortex and Adult Neurogenesis in Major Depression. Int. J. Mol. Sci. 2021, 22, 11725. [Google Scholar] [CrossRef] [PubMed]
  87. Scrivo, R.; Vasile, M.; Bartosiewicz, I.; Valesini, G. Inflammation as “common soil” of the multifactorial diseases. Autoimmun. Rev. 2011, 10, 369–374. [Google Scholar] [CrossRef] [PubMed]
  88. Martone, G. The inflammation hypothesis and mental illness. J. Clin. Psychiatr. Neurosci. 2019, 2, 3–12. [Google Scholar]
  89. Dolsen, M.R.; Prather, A.A.; Lamers, F.; Penninx, B.W. Suicidal ideation and suicide attempts: Associations with sleep duration, insomnia, and inflammation. Psychol. Med. 2021, 51, 2094–2103. [Google Scholar] [CrossRef]
  90. Brahadeeswaran, S.; Sivagurunathan, N.; Calivarathan, L. Inflammasome Signaling in the Aging Brain and Age-Related Neurodegenerative Diseases. Mol. Neurobiol. 2022, 59, 2288–2304. [Google Scholar] [CrossRef] [PubMed]
  91. Capuron, L.; Raison, C.L.; Musselman, D.L.; Lawson, D.H.; Nemeroff, C.B.; Miller, A.H. Association of exaggerated HPA axis response to the initial injection of interferon-alpha with development of depression during interferon-alpha therapy. Am. J. Psychiatr. 2003, 160, 1342–1345. [Google Scholar] [CrossRef]
  92. Davies, K.A.; Cooper, E.; Voon, V.; Tibble, J.; Cercignani, M.; Harrison, N.A. Interferon and anti-TNF therapies differentially modulate amygdala reactivity which predicts associated bidirectional changes in depressive symptoms. Mol. Psychiatr. 2021, 26, 5150–5160. [Google Scholar] [CrossRef]
  93. Su, K.-P.; Lai, H.-C.; Peng, C.-Y.; Su, W.-P.; Chang, J.P.-C.; Pariante, C.M. Interferon-alpha-induced depression: Comparisons between early-and late-onset subgroups and with patients with major depressive disorder. Brain Behav. Immun. 2019, 80, 512–518. [Google Scholar] [CrossRef] [PubMed]
  94. Dooley, L.N.; Kuhlman, K.R.; Robles, T.F.; Eisenberger, N.I.; Craske, M.G.; Bower, J.E. The role of inflammation in core features of depression: Insights from paradigms using exogenously-induced inflammation. Neurosci. Biobehav. Rev. 2018, 94, 219–237. [Google Scholar] [CrossRef] [Green Version]
  95. Ma, K.; Zhang, H.; Baloch, Z. Pathogenetic and therapeutic applications of tumor necrosis factor-α (TNF-α) in major depressive disorder: A systematic review. Int. J. Mol. Sci. 2016, 17, 733. [Google Scholar] [CrossRef] [Green Version]
  96. Hernández-Hernández, O.T.; Martínez-Mota, L.; Herrera-Pérez, J.J.; Jiménez-Rubio, G. Role of estradiol in the expression of genes involved in serotonin neurotransmission: Implications for female depression. Curr. Neuropharmacol. 2019, 17, 459–471. [Google Scholar] [CrossRef] [PubMed]
  97. Baudry, A.; Pietri, M.; Launay, J.-M.; Kellermann, O.; Schneider, B. Multifaceted regulations of the serotonin transporter: Impact on antidepressant response. Front. Neurosci. 2019, 13, 91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Wu, H.; Denna, T.H.; Storkersen, J.N.; Gerriets, V.A. Beyond a neurotransmitter: The role of serotonin in inflammation and immunity. Pharmacol. Res. 2019, 140, 100–114. [Google Scholar] [CrossRef]
  99. Smith, R.S. The macrophage theory of depression. Med. Hypotheses 1991, 35, 298–306. [Google Scholar] [CrossRef]
  100. Norman, G.; Karelina, K.; Zhang, N.; Walton, J.; Morris, J.; Devries, A. Stress and IL-1β contribute to the development of depressive-like behavior following peripheral nerve injury. Mol. Psychiatr. 2010, 15, 404–414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Wang, Y.-L.; Han, Q.-Q.; Gong, W.-Q.; Pan, D.-H.; Wang, L.-Z.; Hu, W.; Yang, M.; Li, B.; Yu, J.; Liu, Q. Microglial activation mediates chronic mild stress-induced depressive-and anxiety-like behavior in adult rats. J. Neuroinflamm. 2018, 15, 198–209. [Google Scholar] [CrossRef] [Green Version]
  102. Miura, H.; Ozaki, N.; Sawada, M.; Isobe, K.; Ohta, T.; Nagatsu, T. A link between stress and depression: Shifts in the balance between the kynurenine and serotonin pathways of tryptophan metabolism and the etiology and pathophysiology of depression. Stress 2008, 11, 198–209. [Google Scholar] [CrossRef]
  103. Tanaka, M.; Tóth, F.; Polyák, H.; Szabó, Á.; Mándi, Y.; Vécsei, L. Immune influencers in action: Metabolites and enzymes of the Tryptophan-Kynurenine metabolic pathway. Biomedicines 2021, 9, 734. [Google Scholar] [CrossRef]
  104. 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. Psychiatr. 2010, 67, 446–457. [Google Scholar] [CrossRef]
  105. Raison, C.; Borisov, A.; Woolwine, B.; Massung, B.; Vogt, G.; Miller, A. Interferon-α effects on diurnal hypothalamic–pituitary–adrenal axis activity: Relationship with proinflammatory cytokines and behavior. Mol. Psychiatr. 2010, 15, 535–547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Bekhbat, M.; Treadway, M.T.; Felger, J.C. Inflammation as a Pathophysiologic Pathway to Anhedonia: Mechanisms and Therapeutic Implications; Springer: Berlin/Heidelberg, Germany, 2022. [Google Scholar]
  107. Bauer, M.E.; Teixeira, A.L. Inflammation in psychiatric disorders: What comes first? Ann. N. Y. Acad. Sci. 2019, 1437, 57–67. [Google Scholar] [CrossRef] [PubMed]
  108. Peng, Y.-L.; Liu, Y.-N.; Liu, L.; Wang, X.; Jiang, C.-L.; Wang, Y.-X. Inducible nitric oxide synthase is involved in the modulation of depressive behaviors induced by unpredictable chronic mild stress. J. Neuroinflamm. 2012, 9, 75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Henry, C.J.; Huang, Y.; Wynne, A.; Hanke, M.; Himler, J.; Bailey, M.T.; Sheridan, J.F.; Godbout, J.P. Minocycline attenuates lipopolysaccharide (LPS)-induced neuroinflammation, sickness behavior, and anhedonia. J. Neuroinflamm. 2008, 5, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Schroder, K.; Tschopp, J. The inflammasomes. Cell 2010, 140, 821–832. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Ketelut-Carneiro, N.; Fitzgerald, K.A. Inflammasomes. Curr. Biol. 2020, 30, R689–R694. [Google Scholar] [CrossRef]
  112. Zhang, Y.; Liu, L.; Peng, Y.L.; Liu, Y.Z.; Wu, T.Y.; Shen, X.L.; Zhou, J.R.; Sun, D.Y.; Huang, A.J.; Wang, X. Involvement of inflammasome activation in lipopolysaccharide-induced mice depressive-like behaviors. CNS Neurosci. Ther. 2014, 20, 119–124. [Google Scholar] [CrossRef]
  113. Zhang, L.; Previn, R.; Lu, L.; Liao, R.-F.; Jin, Y.; Wang, R.-K. Crocin, a natural product attenuates lipopolysaccharide-induced anxiety and depressive-like behaviors through suppressing NF-kB and NLRP3 signaling pathway. Brain Res. Bull. 2018, 142, 352–359. [Google Scholar] [CrossRef]
  114. Wong, M.-L.; Inserra, A.; Lewis, M.; Mastronardi, C.A.; Leong, L.; Choo, J.; Kentish, S.; Xie, P.; Morrison, M.; Wesselingh, S. Inflammasome signaling affects anxiety-and depressive-like behavior and gut microbiome composition. Mol. Psychiatr. 2016, 21, 797–805. [Google Scholar] [CrossRef]
  115. Inserra, A.; Mastronardi, C.A.; Rogers, G.; Licinio, J.; Wong, M.-L. Neuroimmunomodulation in major depressive disorder: Focus on caspase 1, inducible nitric oxide synthase, and interferon-gamma. Mol. Neurobiol. 2019, 56, 4288–4305. [Google Scholar] [CrossRef] [Green Version]
  116. Carlessi, A.S.; Borba, L.A.; Zugno, A.I.; Quevedo, J.; Réus, G.Z. Gut microbiota–brain axis in depression: The role of neuroinflammation. Eur. J. Neurosci. 2021, 53, 222–235. [Google Scholar] [CrossRef] [PubMed]
  117. Ma, Q.; Xing, C.; Long, W.; Wang, H.Y.; Liu, Q.; Wang, R.-F. Impact of microbiota on central nervous system and neurological diseases: The gut-brain axis. J. Neuroinflamm. 2019, 16, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Anderson, R.J.; Freedland, K.E.; Clouse, R.E.; Lustman, P.J. The prevalence of comorbid depression in adults with diabetes: A meta-analysis. Diabetes Care. 2001, 24, 1069–1078. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Ali, S.; Stone, M.; Peters, J.; Davies, M.; Khunti, K. The prevalence of co-morbid depression in adults with Type 2 diabetes: A systematic review and meta-analysis. Diabet. Med. 2006, 23, 1165–1173. [Google Scholar] [CrossRef]
  120. Moulton, C.D.; Pickup, J.C.; Ismail, K. The link between depression and diabetes: The search for shared mechanisms. Lancet Diabetes Endocrinol. 2015, 3, 461–471. [Google Scholar] [CrossRef]
  121. Laake, J.-P.S.; Stahl, D.; Amiel, S.A.; Petrak, F.; Sherwood, R.A.; Pickup, J.C.; Ismail, K. The association between depressive symptoms and systemic inflammation in people with type 2 diabetes: Findings from the South London Diabetes Study. Diabetes Care 2014, 37, 2186–2192. [Google Scholar] [CrossRef] [Green Version]
  122. Zhang, L.; Fu, T.; Yin, R.; Zhang, Q.; Shen, B. Prevalence of depression and anxiety in systemic lupus erythematosus: A systematic review and meta-analysis. BMC Psychiatr. 2017, 17, 1–14. [Google Scholar] [CrossRef] [Green Version]
  123. Van Exel, E.; Jacobs, J.; Korswagen, L.; Voskuyl, A.; Stek, M.; Dekker, J.; Bultink, I. Depression in systemic lupus erythematosus, dependent on or independent of severity of disease. Lupus 2013, 22, 1462–1469. [Google Scholar] [CrossRef]
  124. Xie, X.; Wu, D.; Chen, H. Prevalence and risk factors of anxiety and depression in patients with systemic lupus erythematosus in Southwest China. Rheumatol. Int. 2016, 36, 1705–1710. [Google Scholar] [CrossRef]
  125. Schmeding, A.; Schneider, M. Fatigue, health-related quality of life and other patient-reported outcomes in systemic lupus erythematosus. Best Pract. Res. Clin. Rheumatol. 2013, 27, 363–375. [Google Scholar] [CrossRef]
  126. Mak, A.; Tang, C.; Ho, R.C.-M. Serum tumour necrosis factor-alpha is associated with poor health-related quality of life and depressive symptoms in patients with systemic lupus erythematosus. Lupus 2013, 22, 254–261. [Google Scholar] [CrossRef]
  127. Quan, W.; An, J.; Li, G.; Qian, G.; Jin, M.; Feng, C.; Li, S.; Li, X.; Xu, Y.; Hu, X. Th cytokine profile in childhood-onset systemic lupus erythematosus. BMC Pediatr. 2021, 21, 1–10. [Google Scholar] [CrossRef] [PubMed]
  128. Lin, M.-C.; Guo, H.-R.; Lu, M.-C.; Livneh, H.; Lai, N.-S.; Tsai, T.-Y. Increased risk of depression in patients with rheumatoid arthritis: A seven-year population-based cohort study. Clinics 2015, 70, 91–96. [Google Scholar] [CrossRef]
  129. Matcham, F.; Rayner, L.; Steer, S.; Hotopf, M. The prevalence of depression in rheumatoid arthritis: A systematic review and meta-analysis. Rheumatology 2013, 52, 2136–2148. [Google Scholar] [CrossRef] [Green Version]
  130. Stebbings, S.; Treharne, G.J. Fatigue in rheumatic disease: An overview. Int. J. Clin. Rheumatol. 2010, 5, 487–502. [Google Scholar] [CrossRef]
  131. Kojima, M.; Kojima, T.; Suzuki, S.; Oguchi, T.; Oba, M.; Tsuchiya, H.; Sugiura, F.; Kanayama, Y.; Furukawa, T.A.; Tokudome, S. Depression, inflammation, and pain in patients with rheumatoid arthritis. Arthritis Care Res. 2009, 61, 1018–1024. [Google Scholar] [CrossRef] [PubMed]
  132. Madsen, S.G.; Danneskiold-Samsøe, B.; Stockmarr, A.; Bartels, E. Correlations between fatigue and disease duration, disease activity, and pain in patients with rheumatoid arthritis: A systematic review. Scand. J. Rheumatol. 2016, 45, 255–261. [Google Scholar] [CrossRef] [Green Version]
  133. Almeida, C.; Choy, E.H.; Hewlett, S.; Kirwan, J.R.; Cramp, F.; Chalder, T.; Pollock, J.; Christensen, R. Biologic interventions for fatigue in rheumatoid arthritis. Cochrane Database Syst. Rev. 2016, 2016, CD008334. [Google Scholar] [CrossRef]
  134. Yende, S.; D’Angelo, G.; Kellum, J.A.; Weissfeld, L.; Fine, J.; Welch, R.D.; Kong, L.; Carter, M.; Angus, D.C. Inflammatory markers at hospital discharge predict subsequent mortality after pneumonia and sepsis. Am. J. Respir. Crit. Care Med. 2008, 177, 1242–1247. [Google Scholar] [CrossRef] [Green Version]
  135. Shukla, P.; Rao, G.M.; Pandey, G.; Sharma, S.; Mittapelly, N.; Shegokar, R.; Mishra, P.R. Therapeutic interventions in sepsis: Current and anticipated pharmacological agents. Br. J. Pharmacol. 2014, 171, 5011–5031. [Google Scholar] [CrossRef] [Green Version]
  136. Prescott, H.C.; Angus, D.C. Enhancing recovery from sepsis: A review. JAMA 2018, 319, 62–75. [Google Scholar] [CrossRef] [PubMed]
  137. Davydow, D.S.; Hough, C.L.; Langa, K.M.; Iwashyna, T.J. Symptoms of depression in survivors of severe sepsis: A prospective cohort study of older Americans. Am. J. Geriatr. Psychiatr. 2013, 21, 887–897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Hughes, M.M.; Connor, T.J.; Harkin, A. Stress-related immune markers in depression: Implications for treatment. Int. J. Neuropsychopharmacol. 2016, 19, pyw001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Ojard, C.; Donnelly, J.P.; Safford, M.M.; Griffin, R. Psychosocial stress as a risk factor for sepsis: A population-based cohort study. Psychosom. Med. 2015, 77, 93. [Google Scholar] [CrossRef] [Green Version]
  140. Anderson, S.T.; Commins, S.; Moynagh, P.N.; Coogan, A.N. Lipopolysaccharide-induced sepsis induces long-lasting affective changes in the mouse. Brain Behav. Immun. 2015, 43, 98–109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Cassol-Jr, O.J.; Comim, C.M.; Petronilho, F.; Constantino, L.S.; Streck, E.L.; Quevedo, J.; Dal-Pizzol, F. Low dose dexamethasone reverses depressive-like parameters and memory impairment in rats submitted to sepsis. Neurosci. Lett. 2010, 473, 126–130. [Google Scholar] [CrossRef] [PubMed]
  142. Chiu, W.; Su, Y.; Su, K.; Chen, P. Recurrence of depressive disorders after interferon-induced depression. Transl. Psychiatr. 2017, 7, e1026. [Google Scholar] [CrossRef] [Green Version]
  143. Zhang, G.; Xu, S.; Zhang, Z.; Zhang, Y.; Wu, Y.; An, J.; Lin, J.; Yuan, Z.; Shen, L.; Si, T. Identification of key genes and the pathophysiology associated with major depressive disorder patients based on integrated bioinformatics analysis. Front. Psychiatr. 2020, 11, 192. [Google Scholar] [CrossRef] [Green Version]
  144. Ohgi, Y.; Futamura, T.; Kikuchi, T.; Hashimoto, K. Effects of antidepressants on alternations in serum cytokines and depressive-like behavior in mice after lipopolysaccharide administration. Pharmacol. Biochem. Behav. 2013, 103, 853–859. [Google Scholar] [CrossRef]
  145. Ramirez, K.; Shea, D.T.; McKim, D.B.; Reader, B.F.; Sheridan, J.F. Imipramine attenuates neuroinflammatory signaling and reverses stress-induced social avoidance. Brain Behav. Immun. 2015, 46, 212–220. [Google Scholar] [CrossRef] [Green Version]
  146. Qiu, W.; Wu, M.; Liu, S.; Chen, B.; Pan, C.; Yang, M.; Wang, K.-J. Suppressive immunoregulatory effects of three antidepressants via inhibition of the nuclear factor-κB activation assessed using primary macrophages of carp (Cyprinus carpio). Toxicol. Appl. Pharmacol. 2017, 322, 1–8. [Google Scholar] [CrossRef] [PubMed]
  147. Munzer, A.; Sack, U.; Mergl, R.; Schönherr, J.; Petersein, C.; Bartsch, S.; Kirkby, K.C.; Bauer, K.; Himmerich, H. Impact of antidepressants on cytokine production of depressed patients in vitro. Toxins 2013, 5, 2227–2240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Hannestad, J.; DellaGioia, N.; Bloch, M. The effect of antidepressant medication treatment on serum levels of inflammatory cytokines: A meta-analysis. Neuropsychopharmacology 2011, 36, 2452–2459. [Google Scholar] [CrossRef] [PubMed]
  149. Więdłocha, M.; Marcinowicz, P.; Krupa, R.; Janoska-Jaździk, M.; Janus, M.; Dębowska, W.; Mosiołek, A.; Waszkiewicz, N.; Szulc, A. Effect of antidepressant treatment on peripheral inflammation markers—A meta-analysis. Prog. Neuropsychopharmacol. Biol. Psychiatr. 2018, 80, 217–226. [Google Scholar] [CrossRef] [PubMed]
  150. Chen, C.-Y.; Yeh, Y.-W.; Kuo, S.-C.; Liang, C.-S.; Ho, P.-S.; Huang, C.-C.; Yen, C.-H.; Shyu, J.-F.; Lu, R.-B.; Huang, S.-Y. Differences in immunomodulatory properties between venlafaxine and paroxetine in patients with major depressive disorder. Psychoneuroendocrinology 2018, 87, 108–118. [Google Scholar] [CrossRef] [PubMed]
  151. Mohr, D.C.; Goodkin, D.E.; Islar, J.; Hauser, S.L.; Genain, C.P. Treatment of depression is associated with suppression of nonspecific and antigen-specific TH1 responses in multiple sclerosis. Arch. Neurol. 2001, 58, 1081–1086. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. 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] [Green Version]
  153. Rethorst, C.D.; Toups, M.S.; Greer, T.L.; Nakonezny, P.A.; Carmody, T.J.; Grannemann, B.D.; Huebinger, R.M.; Barber, R.C.; Trivedi, M.H. Pro-inflammatory cytokines as predictors of antidepressant effects of exercise in major depressive disorder. Mol. Psychiatr. 2013, 18, 1119–1124. [Google Scholar] [CrossRef]
  154. Yrondi, A.; Sporer, M.; Peran, P.; Schmitt, L.; Arbus, C.; Sauvaget, A. Electroconvulsive therapy, depression, the immune system and inflammation: A systematic review. Brain Stimul. 2018, 11, 29–51. [Google Scholar] [CrossRef]
  155. Freire, T.F.V.; da Rocha, N.S.; de Almeida Fleck, M.P. The association of electroconvulsive therapy to pharmacological treatment and its influence on cytokines. J. Psychiatr. Res. 2017, 92, 205–211. [Google Scholar] [CrossRef]
  156. Kronfol, Z.; Nair, M.P.; Weinberg, V.; Young, E.A.; Aziz, M. Acute effects of electroconvulsive therapy on lymphocyte natural killer cell activity in patients with major depression. J. Affect Disord. 2002, 71, 211–215. [Google Scholar] [CrossRef]
  157. Kiraly, D.; Horn, S.; Van Dam, N.; Costi, S.; Schwartz, J.; Kim-Schulze, S.; Patel, M.; Hodes, G.E.; Russo, S.; Merad, M. Altered peripheral immune profiles in treatment-resistant depression: Response to ketamine and prediction of treatment outcome. Translational Psychiatr. 2017, 7, e1065. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Myung, W.; Lim, S.-W.; Woo, H.I.; Park, J.H.; Shim, S.; Lee, S.-Y.; Kim, D.K. Serum cytokine levels in major depressive disorder and its role in antidepressant response. Psychiatr. Investig. 2016, 13, 644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Uher, R.; Tansey, K.E.; Dew, T.; Maier, W.; Mors, O.; Hauser, J.; Dernovsek, M.Z.; Henigsberg, N.; Souery, D.; Farmer, A. An inflammatory biomarker as a differential predictor of outcome of depression treatment with escitalopram and nortriptyline. Am. J. Psychiatr. 2014, 171, 1278–1286. [Google Scholar] [CrossRef] [PubMed]
  160. Yoshimura, R.; Hori, H.; Ikenouchi-Sugita, A.; Umene-Nakano, W.; Ueda, N.; Nakamura, J. Higher plasma interleukin-6 (IL-6) level is associated with SSRI-or SNRI-refractory depression. Prog. Neuropsychopharmacol. Biol. Psychiatr. 2009, 33, 722–726. [Google Scholar] [CrossRef]
  161. Eller, T.; Vasar, V.; Shlik, J.; Maron, E. Pro-inflammatory cytokines and treatment response to escitaloprsam in major depressive disorder. Prog. Neuropsychopharmacol. Biol. Psychiatr. 2008, 32, 445–450. [Google Scholar] [CrossRef]
  162. Benedetti, F.; Lucca, A.; Brambilla, F.; Colombo, C.; Smeraldi, E. Interleukine-6 serum levels correlate with response to antidepressant sleep deprivation and sleep phase advance. Prog. Neuropsychopharmacol. Biol. Psychiatr. 2002, 26, 1167–1170. [Google Scholar] [CrossRef]
  163. Sorri, A.; Järventausta, K.; Kampman, O.; Lehtimäki, K.; Björkqvist, M.; Tuohimaa, K.; Hämäläinen, M.; Moilanen, E.; Leinonen, E. Low tumor necrosis factor-α levels predict symptom reduction during electroconvulsive therapy in major depressive disorder. Brain Behav. 2018, 8, e00933. [Google Scholar] [CrossRef]
  164. Yang, T.-T.; Wang, L.; Deng, X.-Y.; Yu, G. Pharmacological treatments for fatigue in patients with multiple sclerosis: A systematic review and meta-analysis. J. Neurol. Sci. 2017, 380, 256–261. [Google Scholar] [CrossRef]
  165. Rogóż, Z.; Kubera, M.; Rogóż, K.; Basta-Kaim, A.; Budziszewska, B. Effect of co-administration of fluoxetine and amantadine on immunoendocrine parameters in rats subjected to a forced swimming test. Pharmacol. Rep. 2009, 61, 1050–1060. [Google Scholar] [CrossRef]
  166. Lin, P.-Y.; Huang, S.-Y.; Su, K.-P. A meta-analytic review of polyunsaturated fatty acid compositions in patients with depression. Biol. Psychiatr. 2010, 68, 140–147. [Google Scholar] [CrossRef] [PubMed]
  167. Appleton, K.M.; Voyias, P.D.; Sallis, H.M.; Dawson, S.; Ness, A.R.; Churchill, R.; Perry, R. Omega-3 fatty acids for depression in adults. Cochrane Database Syst. Rev. 2021, 11, CD004692. [Google Scholar] [PubMed]
  168. Hallahan, B.; Ryan, T.; Hibbeln, J.R.; Murray, I.T.; Glynn, S.; Ramsden, C.E.; SanGiovanni, J.P.; Davis, J.M. Efficacy of omega-3 highly unsaturated fatty acids in the treatment of depression. Br. J. Psychiatr. 2016, 209, 192–201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Luo, X.-D.; Feng, J.-S.; Yang, Z.; Huang, Q.-T.; Lin, J.-D.; Yang, B.; Su, K.-P.; Pan, J.-Y. High-dose omega-3 polyunsaturated fatty acid supplementation might be more superior than low-dose for major depressive disorder in early therapy period: A network meta-analysis. BMC Psychiatr. 2020, 20, 1–8. [Google Scholar] [CrossRef] [PubMed]
  170. Shi, Z.; Ren, H.; Huang, Z.; Peng, Y.; He, B.; Yao, X.; Yuan, T.-F.; Su, H. Fish oil prevents lipopolysaccharide-induced depressive-like behavior by inhibiting neuroinflammation. Mol. Neurobiol. 2017, 54, 7327–7334. [Google Scholar] [CrossRef] [PubMed]
  171. Su, K.-P.; Lai, H.-C.; Yang, H.-T.; Su, W.-P.; Peng, C.-Y.; Chang, J.P.-C.; Chang, H.-C.; Pariante, C.M. Omega-3 fatty acids in the prevention of interferon-alpha-induced depression: Results from a randomized, controlled trial. Biol. Psychiatr. 2014, 76, 559–566. [Google Scholar] [CrossRef]
  172. Schuch, F.B.; Vancampfort, D.; Richards, J.; Rosenbaum, S.; Ward, P.B.; Stubbs, B. Exercise as a treatment for depression: A meta-analysis adjusting for publication bias. J. Psychiatr. Res. 2016, 77, 42–51. [Google Scholar] [CrossRef] [Green Version]
  173. Morres, I.D.; Hatzigeorgiadis, A.; Stathi, A.; Comoutos, N.; Arpin-Cribbie, C.; Krommidas, C.; Theodorakis, Y. Aerobic exercise for adult patients with major depressive disorder in mental health services: A systematic review and meta-analysis. Depress. Anxiety 2019, 36, 39–53. [Google Scholar] [CrossRef] [Green Version]
  174. Wipfli, B.M.; Rethorst, C.D.; Landers, D.M. The anxiolytic effects of exercise: A meta-analysis of randomized trials and dose–response analysis. J. Sport Exerc. Psychol. 2008, 30, 392–410. [Google Scholar] [CrossRef] [Green Version]
  175. Phillips, C.; Fahimi, A. Immune and neuroprotective effects of physical activity on the brain in depression. Front. Neurosci. 2018, 12, 498. [Google Scholar] [CrossRef] [Green Version]
  176. Carrea-Gonzalez, M.D.P.; Canton-Habas, V.; Rich-Ruiz, M. Age, depression and dementia: The inflammatory process. Adv. Clin. Exp. Med. 2022, 31, 469–473. [Google Scholar] [CrossRef] [PubMed]
  177. Chen, C. Recent advances in the study of the comorbidity of depressive and anxiety disorders. Adv. Clin. Exp. Med. 2022, 31, 355–358. [Google Scholar] [CrossRef]
  178. Tanaka, M.; Spekker, E.; Szabó, Á.; Polyák, H.; Vécsei, L. Modelling the neurodevelopmental pathogenesis in neuropsychiatric disorders. Bioactive kynurenines and their analogues as neuroprotective agents-in celebration of 80th birthday of Professor Peter Riederer. J. Neural Transm. 2022, 129, 627–642. [Google Scholar] [CrossRef] [PubMed]
  179. Tanaka, M.; Török, N.; Tóth, F.; Szabó, Á.; Vécsei, L. Co-Players in Chronic Pain: Neuroinflammation and the Tryptophan-Kynurenine Metabolic Pathway. Biomedicines 2021, 9, 897. [Google Scholar] [CrossRef] [PubMed]
  180. Hunt, C.; Macedo ECordeiro, 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]
  181. Sanjay Park, M.; Lee, H.J. Roles of Fatty Acids in Microglial Polarization: Evidence from In Vitro and In Vivo Studies on Neurodegenerative Diseases. Int. J. Mol. Sci. 2022, 23, 7300. [Google Scholar] [CrossRef]
Figure 1. Assumptive model of reciprocal interactions between low-grade inflammatory changes and MDD. Negative health behaviors, including poor stress-coping strategies, may provoke inflammatory responses, which involve potential mechanisms mediating the effects of such negative behaviors on inflammation, dysregulation of n-3 fatty acids, and gut physiology dysregulation. This, in turn, may generate symptoms of MDD, including motivational deprivation and psychomotor impairment, further aggravating sickness behavior patterns, and may induce depression and more negative health behaviors. Abbreviations: omega-3 (n-3), lipopolysaccharide (LPS), mitogen-activated protein kinases (MAPKs), major depressive disorder (MDD).
Figure 1. Assumptive model of reciprocal interactions between low-grade inflammatory changes and MDD. Negative health behaviors, including poor stress-coping strategies, may provoke inflammatory responses, which involve potential mechanisms mediating the effects of such negative behaviors on inflammation, dysregulation of n-3 fatty acids, and gut physiology dysregulation. This, in turn, may generate symptoms of MDD, including motivational deprivation and psychomotor impairment, further aggravating sickness behavior patterns, and may induce depression and more negative health behaviors. Abbreviations: omega-3 (n-3), lipopolysaccharide (LPS), mitogen-activated protein kinases (MAPKs), major depressive disorder (MDD).
Biomedicines 10 01929 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kim, I.-B.; Lee, J.-H.; Park, S.-C. The Relationship between Stress, Inflammation, and Depression. Biomedicines 2022, 10, 1929. https://doi.org/10.3390/biomedicines10081929

AMA Style

Kim I-B, Lee J-H, Park S-C. The Relationship between Stress, Inflammation, and Depression. Biomedicines. 2022; 10(8):1929. https://doi.org/10.3390/biomedicines10081929

Chicago/Turabian Style

Kim, Il-Bin, Jae-Hon Lee, and Seon-Cheol Park. 2022. "The Relationship between Stress, Inflammation, and Depression" Biomedicines 10, no. 8: 1929. https://doi.org/10.3390/biomedicines10081929

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