Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2994 word(s) 2994 2021-10-08 07:53:14 |
2 corrected the format Meta information modification 2994 2022-01-28 02:30:25 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Lorkiewicz, P. Biomarkers of Post-COVID Depression. Encyclopedia. Available online: https://encyclopedia.pub/entry/18911 (accessed on 05 May 2024).
Lorkiewicz P. Biomarkers of Post-COVID Depression. Encyclopedia. Available at: https://encyclopedia.pub/entry/18911. Accessed May 05, 2024.
Lorkiewicz, Piotr. "Biomarkers of Post-COVID Depression" Encyclopedia, https://encyclopedia.pub/entry/18911 (accessed May 05, 2024).
Lorkiewicz, P. (2022, January 27). Biomarkers of Post-COVID Depression. In Encyclopedia. https://encyclopedia.pub/entry/18911
Lorkiewicz, Piotr. "Biomarkers of Post-COVID Depression." Encyclopedia. Web. 27 January, 2022.
Biomarkers of Post-COVID Depression
Edit

Post-COVID depression affects people who have had SARS-CoV-2 infection. A very important issue for the mental health of the general population is to look for the causes of this complication and its biomarkers. This will help in faster diagnosis and effective treatment of the affected patients. 

COVID-19 depression biomarkers post-COVID kynurenine cytokines

1. Introduction

The most common symptoms of coronavirus disease are fever, cough, shortness of breath, muscle pain, headache, diarrhea, rhinorrhea, loss of smell and taste [1][2][3]. In addition, there are more and more reports of mental health problems in people who have survived SARS-CoV-2 infection. The most frequently described mental disorders are major depressive disorder (MDD), post-traumatic stress disorder (PTSD), anxiety disorders, obsessive-compulsive disorders (OCD) and insomnia [4][5][6]. These disorders occur mainly in the acute phase of infection and shortly after it [6][7][8]. While the symptoms of PTSD, anxiety disorders and insomnia gradually disappear, it has been shown that symptoms of MDD persist even in the third month of follow-up [6].

2. Inflammatory Factors

The role of inflammation and inflammatory factors in the development of MDD is well understood [9][10]. It has been proven that patients suffering from inflammatory diseases, such as multiple sclerosis or systemic lupus, who have also been treated with cytokines have a greater chance of developing MDD [11][12][13][14]. COVID-19 is a disease that can cause systemic inflammation and a cytokine storm [15], and is therefore analogous to inflammatory diseases, and may contribute to the development of major depressive disorder (MDD).
Inflammatory cytokines are molecules that mediate the immune response upon activation of the peripheral immune system. Cytokines such as tumor necrosis factor alpha (TNFα), interferon-γ (IFN-γ), interleukin 1 (IL-1) play a role primarily in enhancing the cellular response, while cytokines such as interleukin 6 (IL-6), interleukin 10 (IL-10), interleukin 13 (IL-13) are more associated with the humoral response [9][16]. Cytokines can be produced in the brain by astrocytes and microglia [17][18] or reach it from the periphery due to several mechanisms such as passage through leaky blood-brain barrier (BBB) regions, including the choroid plexus and periventricular organs, active transport through cytokine transport molecules on the endothelium of the brain, transmission of cytokine signals during an infection in the abdominal cavity via afferent nerve fibers such as the vagus nerve, the passage of activated monocytes into the brain from the periphery, or by signals of second-messengers from the BBB endothelial lining which results in overproduction of cytokines [19][20][21].

2.1. IL-6/sIL-6R

Interleukin 6 (IL-6) is a pro-inflammatory pleiotropic cytokine secreted mainly by monocytes and macrophages under the influence of interleukin 1β and TNF-α, but also by astrocytes and microglia. It belongs to the family of proteins that use gp130 as a signal transmitter [22]
It is the cytokine most often described in MDD, and the studies describing its involvement in MDD are mostly consistent with each other [23][24][9][25][26]. According to some studies, high levels of IL-6 correlate with the severity of MDD symptoms in patients who do not respond to treatment [27]. It has been observed that in patients with MDD, increased levels of IL-6 correlated with attention deficit disorder, and one study showed that its increase was prior to the incidence of cognitive impairment in patients with MDD [26].
High levels of IL-6 is widely described in relation to COVID-19, and its level corresponds to the severity of the disease [6][28][29][30][31]. In one study, high levels of sIL-6R were also detected in few COVID-19 (+) patients, but the results are not consistent and more research is needed [31]. Higher levels of IL-6 in tandem with higher levels of sIL-6R lead to increased trans-signaling [32]. Thus, if the severity of COVID-19 correlates with the amount of IL-6, patients with severe course may be at greater risk of developing post-COVID depression.

2.2. IL-10

Interleukin 10 (IL-10) is an anti-inflammatory cytokine produced by Th2, Treg cells and M2 macrophages [33]. In the central nervous system (CNS) it is produced, inter alia, by astroglia and microglia [34]. In the latter, the secretion of IL-10 is augmented by neurotransmitters and damage-associated molecules—glutamate and adenosine respectively [35]. Overall IL-10 production increases with increased levels of IL-6 and TNF-α [36][37]. It exerts its function by binding with IL-10 receptor (IL-10R) which consist of two subunits—IL-10R1 and IL-10R2. The latter is expressed in most cell types, but IL-10R1 is mostly restricted to cells of hematopoietic lineage. Due to the myeloid origins, microglia express both subunits of IL-10R. Unexpectedly, resting astrocytes also express IL-10R1 [35]. IL-10 limits neuroinflammation, promotes production of immunosuppressive transforming growth factor β (TGF-β) by astrocytes and reduces astrogliosis in response to the pathogenic factors [35].
Elevated levels of IL-10 are often reported in people with MDD [38][39][40]. Some researchers also observed that MDD severity is related to increased IL-10 [41][42]. Two meta-analyzes have shown that IL-10 level decreases with effective antidepressant treatment [38][43].
To date, many studies have described an increased level of IL-10 in COVID-19 patients [44][45][46]. In severe cases, the level of IL-10 was higher than in mild cases, and positively correlated with mortality due to COVID-19 [15][45][47][48][49]. Thus, high levels of IL-10 during and after SARS-CoV-2 infection may suggest an increased susceptibility to developing MDD in these patients. It may also be a good indicator for monitoring the treatment of post-COVID depression, due to its decline with antidepressant treatment [38].

2.3. TNF-α/sTNFR1, sTNFR2

Tumor necrosis factor α (TNF-α) is a pro-inflammatory cytokine produced by Th1 lymphocytes and M1 macrophages, and by astroglia and microglia in the brain [23]. It is one of the earliest cytokines released following trauma, infection or exposure to lipopolysaccharide (LPS) [50] and a regulator of pro-inflammatory cytokine production. Its high level induces the production of, among others, CRP, IL-6, IL-1β [50][51][52].
It is found to be one of the most promising markers of major depressive disorder. Its elevated level, along with CRP and IL-6, is most consistently described in studies on MDD biomarkers [53][54][23][55][24][56][57][9][10][58]. It is one of the major cytokines involved in neuroinflammation [59] and acts as an inhibitor of hippocampal neurogenesis [60][61], an inducer of apoptosis [62][63] and negatively affects synaptogenesis, synaptic plasticity and the structure of synaptic membranes [64][65]. It increases the permeability of BBB [66] and significantly affects the production of serotonin through its ability to activate indoleamine 2,3-dioxygenase (IDO) [67][68].
Its level decreases with effective treatment of MDD [69], and its persistent concentration indicates treatment-resistant depression (TRD) [70][71]. It exerts its effects through the TNF-R1 and TNF-R2 receptors on cell membranes. They can be released into the serum and are elevated in MDD [72][73][74].
TNF-α is elevated in most COVID-19 patients and correlates with the severity of the disease [5][6][28][30][75]. COVID-19 (+) patients requiring intensive care unit (ICU) admission have higher TNF-α levels when compared to the patients who do not require treatment on ICU [45][76]. One study also found that while in cases of sepsis and acute respiratory distress syndrome (ARDS) in COVID-19 (−) patients, TNF-α levels normalized rapidly after the primary immune response [77][78], it is consistently elevated in COVID-19 (+) patients [45]. This may result in an increased chance of developing post-COVID depression due to longer exposure to the pro-inflammatory effects of TNF-α.
In addition, in one of the studies, an increase in the soluble TNF-α receptors—sTNFR1 and TNFR2 in the serum was observed with the increase in the severity of the disease, and the highest levels were recorded in people who eventually died from COVID-19 [47]. However, so far, not many studies have studies described changes in TNF-α receptors levels, so the importance of this biomarker requires further research.

2.4. IL-1β

Interleukin 1β (IL-1β) is another pro-inflammatory cytokine secreted by the same cell types as TNF-α and IL-6, i.e., Th1 lymphocytes and M1 macrophages. 
Research on the role of IL-1β in MDD is not consistent. Some studies report a correlation and an increase of this cytokine in depression [79][80], although not all researchers have found such a link [38][81]. The level of IL-1β increases with the increase in BMI [82] and the number of depressive episodes [23], therefore it may cause inaccuracies in research and difficulties in obtaining uniform results. There have also been studies on the proportionality of the level of IL-1β to the severity of MDD symptoms [83] and the fact that it is a risk factor for TRD [84][85].
IL-1β has been reported to be elevated in COVID-19 and correlated with disease symptoms [86][87][88]. Severe patients have significantly elevated levels [89], and one study found high levels of IL-1β persistent in COVID-19 patients up to 4 weeks after symptom onset—similar to IL-6 [90]. However, not all studies have shown an IL-1β elevation among COVID-19 patients [91][92], therefore, the importance of this interleukin in post-COVID depression requires more investigation.

2.5. IFN-γ

Interferons are a superfamily of endogenous pleiotropic cytokines that play a large role in the maintenance of homeostasis and defense against infection. Interferon gamma (IFN-γ) is a pro-inflammatory cytokine belonging to the type II interferon family [93]. It is secreted mainly by natural killer cells (NK) and CD4 + T cells and macrophages [94].
Disturbances in IFN-γ levels have been documented among patients suffering from MDD [95][96]. Its central or peripheral administration causes symptoms of sickness behavior such as anhedonia, memory and social interaction disorders—the same as seen in MDD [95]. IFN-γ has been shown to activate microglia, which contributes to the development of depression [97]. Moreover, IFN-γ largely activates IDO and contributes to the transition of tryptophan to the kynurenine pathway metabolites which are involved in the pathogenesis of MDD [97][98][99][100].
It has been shown that up to 40% of patients treated with interferon for hepatitis C develop symptoms of depression [98][101].
However, reports on the role of IFN-γ are inconsistent. Some studies demonstrated its increase among people with MDD [99][100][102], and some showed no increase or even a decrease [12][93].
For COVID-19, the research also diverges. Many studies have shown an increase in the concentration of IFN-γ [44][50][51][71][93][98][99][103][104][105], however, several studies showed a decrease [15][106][107]. In some studies, the severity of COVID-19 positively correlated with the level of IFN-γ [48][49][90]. Due to divergent research results, there is still need for more comprehensive studies on this biomarker.

2.6. CRP

C-reactive protein (CRP) is an acute-phase protein produced by liver cells in response to injury, infection or inflammation. Its production is induced by IL-6, and IL-1 enhances this effect. During inflammatory diseases, its serum concentration increases by a minimum of 25% [108]. Baseline CRP levels may be influenced by factors such as body weight, sex, age, nicotinism, and lipid levels [109].
In the context of COVID-19, an increase in CRP concentration is also often described [6][46][110][111][112][113], and its concentration, according to some reports, correlates with the severity of COVID-19 [110][111][112][114][115].
One study reported an association of post-COVID depression with inflammatory biomarkers, where higher CRP was observed in COVID-19 (+) depressed patients than in COVID-19 (+) patients without depression [116]. A separate study reported a decrease in baseline CRP levels in COVID-19 (+) patients in whom the severity of depression symptoms decreased compared to COVID-19 (+) patients in whom CRP and depression symptoms were not significantly reduced [117]. Unlike the previous two, the study by Mazza et al. in the third month of follow-up in patients with post-COVID depression showed no association with CRP [28].
The relationship of CRP to COVID-19 (+) is well documented, but due to the inflammatory nature of the disease and therefore an overall increase in CRP, its utility as a marker of post-COVID depression requires further investigation.

2.7. IL-2/sIL-2R

Interleukin 2 (IL-2) is a pro-inflammatory cytokine produced mainly by CD4 + Th cells and to a lesser extent by T CD8+ and NK cells. It is released mainly in response to an antigen [118]. It exerts its action through IL-2 receptors (IL-2R) present mainly on activated T cells. 
Studies of MDD biomarkers in affected patients have shown an increased level of this cytokine as well [119][120], and its higher concentration is observed in atypical rather than melancholic depression [120]. The influence of IL-2 on the occurrence of depression symptoms is evidenced by the development of depressive symptoms in people and animals to whom it was administered [95][121][122][123]. Moreover, the concentration of sIL-2R is also elevated in people with MDD [38][124][125][126].
In COVID-19 (+) patients, concentrations of both IL-2 and sIL-2R are higher than in COVID-19 (−) patients, especially in severe patients [15][47][76][106][127][128][129]. Two studies also found an association of increased levels of IL-2 [47] and sIL-2R with mortality from COVID-19 [129], and in another, a high sIL-2R/lymphocyte ratio proved to be the best indicator of critical disease differentiation [130].
There are no studies describing a direct relationship of IL-2R/sIL-2R with post-COVID depression, but due to the clearly described increase in their levels in people with MDD and in people with COVID-19, it may be a promising marker.

2.8. MCP1/CCL2

Monocyte Chemoattractant Protein-1/Chemokine ligand 2 (MCP-1/CCL2) is a chemokine produced by many types of cells, e.g., endothelium, fibroblasts, macrophages, monocytes, astrocytes and microglia. 
Patients with COVID-19 also show an increased level of this chemokine compared to healthy people, and in a large proportion of cases it is more elevated in people with severe disease compared to patients with a mild form [6][47][76][89][90]. One study also found that MCP-1/CCL2 elevation positively correlated with mortality from COVID-19 [47].
The described pro-depressive effect of MCP-1/CCL2 and its increased level, especially in severe cases of COVID-19, make it relevant to further research this chemokine in terms of the development of post-COVID depression.

2.9. SAA1

Serum amyloid A (SAA1) is an acute phase protein that is mainly produced in the liver as a result of IL-1β and IL-6 action [131]. It affects many aspects of the inflammation cascade. by binding to various receptors such as toll like receptors 2 and 4 (TLR2 and TLR4) and the receptor for advanced glycation end products (RAGE) and CD36 [132]. SAA1 activates the secretion of cytokines such as TNF-α, IL-6, IL-8, IL-23, IL-18 and IL-10 [39]. TLR2 and TLR4 receptors are present on macrophages, microglia and astrocytes [39][133], and their stimulation by SAA1 causes production of inflammatory cytokines, including IL-6 and TNF-α, which may play a role in the development of neuroinflammation, which contributes to the occurrence of depressive symptoms [134].
Compared to healthy subjects, patients with symptoms of MDD have elevated levels of SAA1 [39][135][136][137]. Patients admitted to hospital for COVID-19 also show elevated levels of this protein compared to healthy controls [49][138][139]. Additionally, its high concentration positively correlated with the severity of the disease and mortality due to COVID-19. One study noted that a decrease in SAA1 within two weeks of disease was associated with the prognosis of clinical improvement in patients, while its persistently high concentration was associated with death [138].
The clear association of SAA1 with MDD and its persistent high concentration in severe COVID-19 (+) patients suggests that a closer look at its relationship with post-COVID depression is needed.

2.10. Kynurenine Pathway

Kynurenine is a tryptophan metabolite formed as a result of its transformation under the influence of the enzymes indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO). 
In a metabolomic study of COVID-19 (+) patients, tryptophan metabolism was the major disorder detected [29]. In patients infected with SARS-CoV-2, decreased levels of tryptophan and serotonin as well as increased levels of kynurenine, 3-hydroxykynurenine, kynurenic acid and picolinic acid (also one of the metabolites of the kynurenine pathway) were found [29][104][140][141]. One of the reasons for the decreased level of tryptophan in COVID-19 (+) patients may also be its decreased absorption in the gut. SARS-CoV-2 causes the internalization and downregulation of ACE2, which is highly expressed in the intestines [142], and which is needed for the expression of the neutral amino acid transporter in the intestinal lumen—B0AT1 [143]. Activation of the kynurenine pathway can be indirectly assessed by the kynurenine to tryptophan ratio, which was significantly higher in COVID-19 (+) patients than in healthy controls and correlated positively with disease severity [29][140][141].
In conclusion, disturbances in tryptophan metabolism and activation of the kynurenine pathway are well described in research on the pathophysiology of MDD and are of great importance in the search for its biomarkers. Analogous changes can be seen in patients infected with SARS-CoV-2 (Figure 2), and at present disturbances in tryptophan metabolism are one of the most promising theories on the development of post-COVID depression [144]. Therefore, it is worth considering metabolites of the kynurenine pathway in future studies for biomarkers of depression developing in patients after SARS-CoV-2 infection, especially in those with a severe course of the disease.
Figure 2. Summary of the most common biomarkers of activated kynurenine pathway in MDD. Note that biomarkers inside dark-green circle coincide with markers of disturbed tryptophan metabolism in COVID-19. Abbreviations: ↑—increased concentration; ↓—decreased concentration; 3-HK—3-hydroxykynurenine; 5-HT—serotonin; KYN—kynurenine; KYNA—kynurenic acid; MDD—major depressive disorder; QA—quinolinic acid; TRP—tryptophan.

3. Growth Factors

3.1. BDNF

Brain derived neurotrophic factor (BDNF) belongs to the family of neuronal growth factors and its physiological role is to support the differentiation, maturation and survival of dopaminergic, cholinergic and serotonergic neurons of the central nervous system [145]. It also exhibits neuroprotective abilities, is involved in neuroplasticity and enhances long-term potentiation [146][147][148]. It is produced by neurons as well as by peripheral cells such as leukocytes and endothelial cells and is able to pass through the BBB [149]. Its decreased level is a common finding in people suffering from MDD, and effective antidepressant therapy restores it to normal levels [149][150][151].
It is proved that ACE2 is associated with a reduction in BDNF levels [152]. It is widely believed that SARS-CoV-2, by using ACE2 to enter cells, causes its downregulation [153]. This mechanism may cause a secondary reduction in BDNF levels. The confirmation of this theory may be reflected in one of the studies performed, in which patients suffering from COVID-19 were tested for serum BDNF levels. The researchers demonstrated that patients with moderate and severe disease have lower BDNF levels than those with mild disease, and during patients’ recovery, their levels returned to normal [110].
The association of growth factors, especially BDNF, with MDD is often indicated in the literature (Figure 3), and the likely mechanism by which SARS-CoV-2 could reduce it may prove it to be a good biomarker of post-COVID depression. Unfortunately, so far only one study mentions the relationship between BDNF and COVID-19, but its results are promising and it is worth doing more research in this direction.
Figure 3. Summary of the most common changes in growth factors in MDD. Note that biomarkers inside dark-yellow circle coincide with changes in growth factors in COVID-19. Abbreviations: ↑—increased concentration; ↓—decreased concentration; BDNF—brain derived neurotrophic factor; FGF-2—fibroblast growth factor-2; IGF-1—insulin-like growth factor-1; NGF—nerve growth factor; VEGF—vascular endothelial growth factor.

References

  1. Grant, M.C.; Geoghegan, L.; Arbyn, M.; Mohammed, Z.; McGuinness, L.; Clarke, E.L.; Wade, R.G. The prevalence of symptoms in 24,410 adults infected by the novel coronavirus (SARS-CoV-2; COVID-19): A systematic review and meta-analysis of 148 studies from 9 countries. PLoS ONE 2020, 15, e0234765.
  2. Vaira, L.A.; Salzano, G.; Deiana, G.; De Riu, G. Anosmia and Ageusia: Common Findings in COVID-19 Patients. Laryngoscope 2020, 130, 1787.
  3. Lai, C.-C.; Shih, T.-P.; Ko, W.-C.; Tang, H.-J.; Hsueh, P.-R. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and coronavirus disease-2019 (COVID-19): The epidemic and the challenges. Int. J. Antimicrob. Agents 2020, 55, 105924.
  4. Steardo, L.; Steardo, L.; Verkhratsky, A. Psychiatric face of COVID-19. Transl. Psychiatry 2020, 10, 261.
  5. Kong, X.; Zheng, K.; Tang, M.; Kong, F.; Zhou, J.; Diao, L.; Wu, S.; Jiao, P.; Su, T.; Dong, Y. Prevalence and Factors Associated with Depression and Anxiety of Hospitalized Patients with COVID-19. medRxiv 2020.
  6. Mazza, M.G.; De Lorenzo, R.; Conte, C.; Poletti, S.; Vai, B.; Bollettini, I.; Melloni, E.M.T.; Furlan, R.; Ciceri, F.; Rovere-Querini, P.; et al. Anxiety and depression in COVID-19 survivors: Role of inflammatory and clinical predictors. Brain Behav. Immun. 2020, 89, 594–600.
  7. De Lorenzo, R.; Conte, C.; Lanzani, C.; Benedetti, F.; Roveri, L.; Mazza, M.G.; Brioni, E.; Giacalone, G.; Canti, V.; Sofia, V.; et al. Residual clinical damage after COVID-19: A retrospective and prospective observational cohort study. PLoS ONE 2020, 15, e0239570.
  8. Bo, H.-X.; Li, W.; Yang, Y.; Wang, Y.; Zhang, Q.; Cheung, T.; Wu, X.; Xiang, Y.-T. Posttraumatic stress symptoms and attitude toward crisis mental health services among clinically stable patients with COVID-19 in China. Psychol. Med. 2020, 51, 1052–1053.
  9. Han, Q.-Q.; Yu, J. Inflammation: A mechanism of depression? Neurosci. Bull. 2014, 30, 515–523.
  10. Amodeo, G.; Trusso, M.A.; Fagiolini, A. Depression and Inflammation: Disentangling a Clear Yet Complex and Multifaceted Link. Neuropsychiatry 2018, 7, 448–457.
  11. Raison, C.L.; Borisov, A.; Broadwell, S.D.; Capuron, L.; Woolwine, B.J.; Jacobson, I.M.; Nemeroff, C.B.; Miller, A.H. Depression During Pegylated Interferon-Alpha Plus Ribavirin Therapy. J. Clin. Psychiatry 2005, 66, 41–48.
  12. Himmerich, H.; Patsalos, O.; Lichtblau, N.; Ibrahim, M.; Dalton, B. Cytokine Research in Depression: Principles, Challenges, and Open Questions. Front. Psychiatry 2019, 10, 30.
  13. Palagini, L.; Mosca, M.; Tani, C.; Gemignani, A.; Mauri, M.; Bombardieri, S. Depression and systemic lupus erythematosus: A systematic review. Lupus 2013, 22, 409–416.
  14. Fermo, S.L.; Barone, R.; Patti, F.; Laisa, P.; Cavallaro, T.L.; Nicoletti, A.; Zappia, M. Outcome of psychiatric symptoms presenting at onset of multiple sclerosis: A retrospective study. Mult. Scler. J. 2010, 16, 742–748.
  15. Pedersen, S.F.; Ho, Y.-C. SARS-CoV-2: A storm is raging. J. Clin. Investig. 2020, 130, 2202–2205.
  16. Lucey, D.R.; Clerici, M.; Shearer, G.M. Type 1 and type 2 cytokine dysregulation in human infectious, neoplastic, and inflammatory diseases. Clin. Microbiol. Rev. 1996, 9, 532.
  17. Smith, J.A.; Das, A.; Ray, S.K.; Banik, N.L. Role of pro-inflammatory cytokines released from microglia in neurodegenerative diseases. Brain Res. Bull. 2012, 87, 10–20.
  18. Choi, S.S.; Lee, H.J.; Lim, I.; Satoh, J.-I.; Kim, S.U. Human Astrocytes: Secretome Profiles of Cytokines and Chemokines. PLoS ONE 2014, 9, e92325.
  19. Watkins, L.R.; Goehler, L.E.; Relton, J.K.; Tartaglia, N.; Silbert, L.; Martin, D.; Maier, S.F. Blockade of interleukin-1 induced hyperthermia by subdiaphragmatic vagotomy: Evidence for vagal mediation of immune-brain communication. Neurosci. Lett. 1995, 183, 27–31.
  20. Quan, N.; Banks, W.A. Brain-immune communication pathways. Brain Behav. Immun. 2007, 21, 727–735.
  21. Plotkin, S.R.; BanksP, W.A.; Kastin, A.J. Comparison of saturable transport and extracellular pathways in the passage of interleukin-1 α across the blood-brain barrier. J. Neuroimmunol. 1996, 67, 41–47.
  22. Tanaka, T.; Narazaki, M.; Kishimoto, T. IL-6 in Inflammation, Immunity, and Disease. Cold Spring Harb. Perspect. Biol. 2014, 6, a016295.
  23. Nobis, A.; Zalewski, D.; Waszkiewicz, N. Peripheral Markers of Depression. J. Clin. Med. 2020, 9, 3793.
  24. Hacimusalar, Y.; Esel, E. Suggested Biomarkers for Major Depressive Disorder. Arch. Neuropsychiatry 2017, 55, 280–290.
  25. Money, K.M.; Olah, Z.; Korade, Z.; Garbett, K.A.; Shelton, R.C.; Mirnics, K. An altered peripheral IL6 response in major depressive disorder. Neurobiol. Dis. 2016, 89, 46–54.
  26. Ting, E.Y.-C.; Yang, A.C.; Tsai, S.-J. Role of Interleukin-6 in Depressive Disorder. Int. J. Mol. Sci. 2020, 21, 2194.
  27. Lanquillon, S.; Krieg, J.-C.; Bening-Abu-Shach, U.; Vedder, H. Cytokine Production and Treatment Response in Major Depressive Disorder. Neuropsychopharmacology 2000, 22, 370–379.
  28. Mazza, M.G.; Palladini, M.; De Lorenzo, R.; Magnaghi, C.; Poletti, S.; Furlan, R.; Ciceri, F.; Rovere-Querini, P.; Benedetti, F. Persistent psychopathology and neurocognitive impairment in COVID-19 survivors: Effect of inflammatory biomarkers at three-month follow-up. Brain Behav. Immun. 2021, 94, 138–147.
  29. Thomas, T.; Stefanoni, D.; Reisz, J.A.; Nemkov, T.; Bertolone, L.; Francis, R.O.; Hudson, K.E.; Zimring, J.C.; Hansen, K.C.; Hod, E.A.; et al. COVID-19 infection alters kynurenine and fatty acid metabolism, correlating with IL-6 levels and renal status. JCI Insight 2020, 5.
  30. Raony, Í.; Figueiredo, C.S.; Pandolfo, P.; Giestal-De-Araujo, E.; Bomfim, P.O.-S.; Savino, W. Psycho-Neuroendocrine-Immune Interactions in COVID-19: Potential Impacts on Mental Health. Front. Immunol. 2020, 11, 1170.
  31. Di Spigna, G.; Cernia, D.S.; Vargas, M.; Buonavolontà, L.; Servillo, G.; Postiglione, L. Drastically elevated levels of Interleukin-6 and its soluble receptor complex in COVID-19 patients with acute respiratory distress. Clin. Med. Investig. 2020, 5.
  32. Anderson, G.; Kubera, M.; Duda, W.; Lasoń, W.; Berk, M.; Maes, M. Increased IL-6 trans-signaling in depression: Focus on the tryptophan catabolite pathway, melatonin and neuroprogression. Pharmacol. Rep. 2013, 65, 1647–1654.
  33. Saraiva, M.; O’Garra, A. The regulation of IL-10 production by immune cells. Nat. Rev. Immunol. 2010, 10, 170–181.
  34. Villacampa, N.; Almolda, B.; Vilella, A.; Campbell, I.L.; González, B.; Castellano, B. Astrocyte-targeted production of IL-10 induces changes in microglial reactivity and reduces motor neuron death after facial nerve axotomy. Glia 2015, 63, 1166–1184.
  35. Burmeister, A.R.; Marriott, I. The Interleukin-10 Family of Cytokines and Their Role in the CNS. Front. Cell. Neurosci. 2018, 12, 458.
  36. Daftarian, P.M.; Kumar, A.; Kryworuchko, M.; Diaz-Mitoma, F. IL-10 production is enhanced in human T cells by IL-12 and IL-6 and in monocytes by tumor necrosis factor-alpha. J. Immunol. 1996, 157, 12–20.
  37. Jin, J.-O.; Han, X.; Yu, Q. Interleukin-6 induces the generation of IL-10-producing Tr1 cells and suppresses autoimmune tissue inflammation. J. Autoimmun. 2013, 40, 28–44.
  38. Köhler, C.A.; Freitas, T.H.; Maes, M.; De Andrade, N.Q.; Liu, C.S.; Fernandes, B.; 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.
  39. Bryleva, E.Y.; Keaton, S.A.; Grit, J.; Madaj, Z.; Smart, L.; Halstead, S.; Achtyes, E.; Brundin, L.; Sauro-Nagendra, A. The acute-phase mediator serum amyloid A is associated with symptoms of depression and fatigue. Acta Psychiatr. Scand. 2017, 135, 409–418.
  40. Maes, M.; Carvalho, A.F. The Compensatory Immune-Regulatory Reflex System (CIRS) in Depression and Bipolar Disorder. Mol. Neurobiol. 2018, 55, 8885–8903.
  41. Iacob, E.; Light, K.C.; Tadler, S.C.; Weeks, H.R.; White, A.T.; Hughen, R.W.; VanHaitsma, T.A.; Bushnell, L.; Light, A.R. Dysregulation of leukocyte gene expression in women with medication-refractory depression versus healthy non-depressed controls. BMC Psychiatry 2013, 13, 273.
  42. Anjum, S.; Qusar, M.M.A.S.; Shahriar, M.; Islam, S.M.A.; Bhuiyan, M.A.; Islam, R. Altered serum interleukin-7 and interleukin-10 are associated with drug-free major depressive disorder. Ther. Adv. Psychopharmacol. 2020, 10.
  43. Hiles, S.A.; Baker, A.L.; de Malmanche, T.; Attia, J. Interleukin-6, C-reactive protein and interleukin-10 after antidepressant treatment in people with depression: A meta-analysis. Psychol. Med. 2012, 42, 2015–2026.
  44. Perlmutter, A. Immunological Interfaces: The COVID-19 Pandemic and Depression. Front. Neurol. 2021, 12, 657004.
  45. Fraser, D.D.; Cepinskas, G.; Slessarev, M.; Martin, C.; Daley, M.; Miller, M.R.; O’Gorman, D.B.; Gill, S.E.; Patterson, E.K.; dos Santos, C.C. Inflammation Profiling of Critically Ill Coronavirus Disease 2019 Patients. Crit. Care Explor. 2020, 2, e0144.
  46. Samprathi, M.; Jayashree, M. Biomarkers in COVID-19: An Up-To-Date Review. Front. Pediatr. 2021, 8, 607647.
  47. Abers, M.S.; Delmonte, O.M.; Ricotta, E.E.; Fintzi, J.; Fink, D.L.; de Jesus, A.A.A.; Zarember, K.A.; Alehashemi, S.; Oikonomou, V.; Desai, J.V.; et al. An immune-based biomarker signature is associated with mortality in COVID-19 patients. JCI Insight 2021, 6.
  48. Zhu, Z.; Cai, T.; Fan, L.; Lou, K.; Hua, X.; Huang, Z.; Gao, G. Clinical value of immune-inflammatory parameters to assess the severity of coronavirus disease 2019. Int. J. Infect. Dis. 2020, 95, 332–339.
  49. Liu, J.; Li, S.; Liu, J.; Liang, B.; Wang, X.; Wang, H.; Li, W.; Tong, Q.; Yi, J.; Zhao, L.; et al. Longitudinal characteristics of lymphocyte responses and cytokine profiles in the peripheral blood of SARS-CoV-2 infected patients. EBioMedicine 2020, 55, 102763.
  50. Parameswaran, N.; Patial, S. Tumor Necrosis Factor-α Signaling in Macrophages. Crit. Rev. Eukaryot. Gene Expr. 2010, 20, 87–103.
  51. Vassalli, P. The Pathophysiology of Tumor Necrosis Factors. Annu. Rev. Immunol. 1992, 10, 411–452.
  52. Maini, R.N.; Elliott, M.J.; Brennan, F.M.; Feldmann, M. Beneficial effects of tumour necrosis factor-alpha (TNF-α) blockade in rheumatoid arthritis (RA). Clin. Exp. Immunol. 1995, 101, 207–212.
  53. Miller, A.H.; Raison, C.L. The role of inflammation in depression: From evolutionary imperative to modern treatment target. Nat. Rev. Immunol. 2015, 16, 22–34.
  54. Lee, C.-H.; Giuliani, F. The Role of Inflammation in Depression and Fatigue. Front. Immunol. 2019, 10, 1696.
  55. Erjavec, G.N.; Sagud, M.; Perkovic, M.N.; Strac, D.S.; Konjevod, M.; Tudor, L.; Uzun, S.; Pivac, N. Depression: Biological markers and treatment. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2020, 105, 110139.
  56. Malik, S.; Singh, R.; Arora, G.; Dangol, A.; Goyal, S. Biomarkers of Major Depressive Disorder: Knowing is Half the Battle. Clin. Psychopharmacol. Neurosci. 2021, 19, 12–25.
  57. Strawbridge, R.; Young, A.H.; Cleare, A.J. Biomarkers for depression: Recent insights, current challenges and future prospects. Neuropsychiatr. Dis. Treat. 2017, 13, 1245–1262.
  58. 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.
  59. Muhammad, M. Tumor Necrosis Factor Alpha: A Major Cytokine of Brain Neuroinflammation. Cytokines 2020.
  60. Monje, M.L.; Toda, H.; Palmer, T.D. Inflammatory Blockade Restores Adult Hippocampal Neurogenesis. Science 2003, 302, 1760–1765.
  61. Iosif, R.E.; Ekdahl, C.T.; Ahlenius, H.; Pronk, C.J.H.; Bonde, S.; Kokaia, Z.; Jacobsen, S.E.W.; Lindvall, O. Tumor Necrosis Factor Receptor 1 Is a Negative Regulator of Progenitor Proliferation in Adult Hippocampal Neurogenesis. J. Neurosci. 2006, 26, 9703–9712.
  62. Rath, P.C.; Aggarwal, B.B. TNF-Induced Signaling in Apoptosis. J. Clin. Immunol. 1999, 19, 350–364.
  63. Buntinx, M.; Moreels, M.; Vandenabeele, F.; Lambrichts, I.; Raus, J.; Steels, P.; Stinissen, P.; Ameloot, M. Cytokine-induced cell death in human oligodendroglial cell lines: I. Synergistic effects of IFN-γ and TNF-α on apoptosis. J. Neurosci. Res. 2004, 76, 834–845.
  64. Stellwagen, D.; Malenka, R.C. Synaptic scaling mediated by glial TNF-α. Nature 2006, 440, 1054–1059.
  65. Sunico, C.R.; Portillo, F.; González-Forero, D.; Moreno-López, B. Nitric Oxide-Directed Synaptic Remodeling in the Adult Mammal CNS. J. Neurosci. 2005, 25, 1448–1458.
  66. Cheng, Y.; Desse, S.; Martinez, A.; Worthen, R.; Jope, R.S.; Beurel, E. TNFα disrupts blood brain barrier integrity to maintain prolonged depressive-like behavior in mice. Brain Behav. Immun. 2018, 69, 556–567.
  67. Dantzer, R. Role of the Kynurenine Metabolism Pathway in Inflammation-Induced Depression: Preclinical Approaches; Springer: Cham, Switzerland, 2016; Volume 31, pp. 117–138.
  68. O’Connor, J.C.; André, C.; Wang, Y.; Lawson, M.A.; Szegedi, S.S.; Lestage, J.; Castanon, N.; Kelley, K.W.; Dantzer, R. Interferon- and Tumor Necrosis Factor- Mediate the Upregulation of Indoleamine 2,3-Dioxygenase and the Induction of Depressive-Like Behavior in Mice in Response to Bacillus Calmette-Guerin. J. Neurosci. 2009, 29, 4200–4209.
  69. Kãhler, C.A.; Freitas, T.H.; Stubbs, B.; Maes, M.; Solmi, M.; Veronese, N.; De Andrade, N.Q.; Morris, G.; Fernandes, B.; Brunoni, A.R.; et al. Peripheral Alterations in Cytokine and Chemokine Levels After Antidepressant Drug Treatment for Major Depressive Disorder: Systematic Review and Meta-Analysis. Mol. Neurobiol. 2017, 55, 1–12.
  70. O’Brien, S.M.; Scully, P.; Fitzgerald, P.; Scott, L.V.; Dinan, T.G. Plasma cytokine profiles in depressed patients who fail to respond to selective serotonin reuptake inhibitor therapy. J. Psychiatr. Res. 2007, 41, 326–331.
  71. 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.
  72. Sowa-Kućma, M.; Styczeń, K.; Siwek, M.; Misztak, P.; Nowak, R.J.; Dudek, D.; Rybakowski, J.K.; Nowak, G.; Maes, M. Lipid Peroxidation and Immune Biomarkers Are Associated with Major Depression and Its Phenotypes, Including Treatment-Resistant Depression and Melancholia. Neurotox. Res. 2017, 33, 448–460.
  73. Teixeira, A.L.; De Souza, R.T.; Zanetti, M.V.; Brunoni, A.R.; Busatto, G.F.; Zarate, C.A., Jr.; Gattaz, W.F.; Machado-Vieira, R. Increased plasma levels of soluble TNF receptors 1 and 2 in bipolar depression and impact of lithium treatment. Hum. Psychopharmacol. Clin. Exp. 2015, 30, 52–56.
  74. Diniz, B.S.; Teixeira, A.L.; Talib, L.L.; Mendonça, V.A.; Gattaz, W.F.; Forlenza, O.V. Increased soluble TNF receptor 2 in antidepressant-free patients with late-life depression. J. Psychiatr. Res. 2010, 44, 917–920.
  75. Fraser, D.D.; Cepinskas, G.; Patterson, E.K.; Slessarev, M.; Martin, C.; Daley, M.; Patel, M.A.; Miller, M.R.; O’Gorman, D.B.; Gill, S.E.; et al. Novel Outcome Biomarkers Identified With Targeted Proteomic Analyses of Plasma From Critically Ill Coronavirus Disease 2019 Patients. Crit. Care Explor. 2020, 2, e0189.
  76. Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506.
  77. Abraham, E.; Anzueto, A.; Gutierrez, G.; Tessler, S.; Pedro, G.S.; Wunderink, R.; Nogare, A.D.; Nasraway, S.; Berman, S.; Cooney, R.; et al. Double-blind randomised controlled trial of monoclonal antibody to human tumour necrosis factor in treatment of septic shock. Lancet 1998, 351, 929–933.
  78. Pinsky, M.R.; Vincent, J.-L.; Deviere, J.; Alegre, M.; Kahn, R.J.; Dupont, E. Serum Cytokine Levels in Human Septic Shock. Chest 1993, 103, 565–575.
  79. Howren, M.B.; Lamkin, D.M.; Suls, J. Associations of Depression with C-Reactive Protein, IL-1, and IL-6: A Meta-Analysis. Psychosom. Med. 2009, 71, 171–186.
  80. Raison, C.L.; Capuron, L.; Miller, A.H. Cytokines sing the blues: Inflammation and the pathogenesis of depression. Trends Immunol. 2006, 27, 24–31.
  81. Haapakoski, R.; Mathieu, J.; Ebmeier, K.P.; Alenius, H.; Kivimäki, M. Cumulative meta-analysis of interleukins 6 and 1β, tumour necrosis factor α and C-reactive protein in patients with major depressive disorder. Brain Behav. Immun. 2015, 49, 206–215.
  82. Ballak, D.B.; Stienstra, R.; Tack, C.J.; Dinarello, C.A.; van Diepen, J.A. IL-1 family members in the pathogenesis and treatment of metabolic disease: Focus on adipose tissue inflammation and insulin resistance. Cytokine 2015, 75, 280–290.
  83. 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.
  84. Uint, L.; Bastos, G.M.; Thurow, H.S.; Borges, J.B.; Hirata, T.D.C.; França, J.I.D.; Hirata, M.H.; Sousa, A.G.D.M.R. Increased levels of plasma IL-1b and BDNF can predict resistant depression patients. Rev. Assoc. Med. Bras. 2019, 65, 361–369.
  85. Cattaneo, A.; Ferrari, C.; Uher, R.; Bocchio-Chiavetto, L.; Riva, M.A.; Pariante, C.M.; The MRC ImmunoPsychiatry Consortium. Absolute Measurements of Macrophage Migration Inhibitory Factor and Interleukin-1-β mRNA Levels Accurately Predict Treatment Response in Depressed Patients. Int. J. Neuropsychopharmacol. 2016, 19.
  86. Liu, B.; Li, M.; Zhou, Z.; Guan, X.; Xiang, Y. Can we use interleukin-6 (IL-6) blockade for coronavirus disease 2019 (COVID-19)-induced cytokine release syndrome (CRS)? J. Autoimmun. 2020, 111, 102452.
  87. Ye, Q.; Wang, B.; Mao, J. The pathogenesis and treatment of the ‘Cytokine Storm’ in COVID-19. J. Infect. 2020, 80, 607–613.
  88. Mehta, P.; McAuley, D.; Brown, M.; Sanchez, E.; Tattersall, R.S.; Manson, J.J. COVID-19: Consider cytokine storm syndromes and immunosuppression. Lancet 2020, 395, 1033–1034.
  89. Catanzaro, M.; Fagiani, F.; Racchi, M.; Corsini, E.; Govoni, S.; Lanni, C. Immune response in COVID-19: Addressing a pharmacological challenge by targeting pathways triggered by SARS-CoV-2. Signal Transduct. Target. Ther. 2020, 5, 1–10.
  90. Zhao, Y.; Qin, L.; Zhang, P.; Li, K.; Liang, L.; Sun, J.; Xu, B.; Dai, Y.; Li, X.; Zhang, C.; et al. Longitudinal COVID-19 profiling associates IL-1RA and IL-10 with disease severity and RANTES with mild disease. JCI Insight 2020, 5, e139834.
  91. Lucas, C.; Wong, P.; Klein, J.; Castro, T.B.R.; Silva, J.; Sundaram, M.; Ellingson, M.K.; Mao, T.; Oh, J.E.; Israelow, B.; et al. Longitudinal analyses reveal immunological misfiring in severe COVID-19. Nature 2020, 584, 463–469.
  92. Del Valle, D.M.; Kim-Schulze, S.; Huang, H.-H.; Beckmann, N.D.; Nirenberg, S.; Wang, B.; Lavin, Y.; Swartz, T.H.; Madduri, D.; Stock, A.; et al. An inflammatory cytokine signature predicts COVID-19 severity and survival. Nat. Med. 2020, 26, 1636–1643.
  93. Pinto, E.F.; Andrade, C. Interferon-Related Depression: A Primer on Mechanisms, Treatment, and Prevention of a Common Clinical Problem. Curr. Neuropharmacol. 2016, 14, 743–748.
  94. Daria, S.; Proma, M.A.; Shahriar, M.; Islam, S.M.A.; Bhuiyan, M.A.; Islam, R. Serum interferon-gamma level is associated with drug-naïve major depressive disorder. SAGE Open Med. 2020, 8, 2050312120974169.
  95. Maes, M.; Berk, M.; Goehler, L.; Song, C.; Anderson, G.; Gałecki, P.; Leonard, B. Depression and sickness behavior are Janus-faced responses to shared inflammatory pathways. BMC Med. 2012, 10, 66.
  96. Maes, M.; Bosmans, E.; De Jongh, R.; Kenisbc, G.; Vandoolaeghea, 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.
  97. Müller, N.; Myint, A.M.; Schwarz, M.J. The impact of neuroimmune dysregulation on neuroprotection and neurotoxicity in psychiatric disorders--relation to drug treatment. Dialog.-Clin. Neurosci. 2009, 11, 319–332.
  98. Bonaccorso, S.; Marino, V.; Puzella, A.; Pasquini, M.; Biondi, M.; Artini, M.; Almerighi, C.; Verkerk, R.; Meltzer, H.; Maes, M. Increased Depressive Ratings in Patients With Hepatitis C Receiving Interferon-α–Based Immunotherapy Are Related to Interferon-α–Induced Changes in the Serotonergic System. J. Clin. Psychopharmacol. 2002, 22, 86–90.
  99. Myint, A.-M.; Leonard, B.E.; Steinbusch, H.W.; Kim, Y.-K. Th1, Th2, and Th3 cytokine alterations in major depression. J. Affect. Disord. 2005, 88, 167–173.
  100. Maes, M.; Scharpé, S.; Meltzer, H.Y.; Okayli, G.; Bosmans, E.; D’Hondt, P.; Bossche, B.V.; Cosyns, P. Increased neopterin and interferon-gamma secretion and lower availability of L-tryptophan in major depression: Further evidence for an immune response. Psychiatry Res. 1994, 54, 143–160.
  101. Udina, M.; Castellvi, P.; Moreno-España, J.; Navinés, R.; Valdés, M.; Forns, X.; Langohr, K.; Solà, R.; Vieta, E.; Martín-Santos, R. Interferon-Induced Depression in Chronic Hepatitis C. J. Clin. Psychiatry 2012, 73, 1128–1138.
  102. Schmidt, F.M.; Lichtblau, N.; Minkwitz, J.; Chittka, T.; Thormann, J.; Kirkby, K.C.; Sander, C.; Mergl, R.; Faßhauer, M.; Stumvoll, M.; et al. Cytokine levels in depressed and non-depressed subjects, and masking effects of obesity. J. Psychiatr. Res. 2014, 55, 29–34.
  103. De Biasi, S.; Meschiari, M.; Gibellini, L.; Bellinazzi, C.; Borella, R.; Fidanza, L.; Gozzi, L.; Iannone, A.; Tartaro, D.L.; Mattioli, M.; et al. Marked T cell activation, senescence, exhaustion and skewing towards TH17 in patients with COVID-19 pneumonia. Nat. Commun. 2020, 11, 1–17.
  104. Fraser, D.D.; Slessarev, M.; Martin, C.M.; Daley, M.; Patel, M.A.; Miller, M.R.; Patterson, E.K.; O’Gorman, D.B.; Gill, S.E.; Wishart, D.S.; et al. Metabolomics Profiling of Critically Ill Coronavirus Disease 2019 Patients: Identification of Diagnostic and Prognostic Biomarkers. Crit. Care Explor. 2020, 2, e0272.
  105. Coperchini, F.; Chiovato, L.; Croce, L.; Magri, F.; Rotondi, M. The cytokine storm in COVID-19: An overview of the involvement of the chemokine/chemokine-receptor system. Cytokine Growth Factor Rev. 2020, 53, 25–32.
  106. Chen, G.; Wu, D.; Guo, W.; Cao, Y.; Huang, D.; Wang, H.; Wang, T.; Zhang, X.; Chen, H.; Yu, H.; et al. Clinical and immunological features of severe and moderate coronavirus disease. J. Clin. Investig. 2020, 130, 2620–2629.
  107. Zheng, H.-Y.; Zhang, M.; Yang, C.-X.; Zhang, N.; Wang, X.-C.; Yang, X.-P.; Dong, X.-Q.; Zheng, Y.-T. Elevated exhaustion levels and reduced functional diversity of T cells in peripheral blood may predict severe progression in COVID-19 patients. Cell. Mol. Immunol. 2020, 17, 541–543.
  108. Du Clos, T.W.; Mold, C. C-Reactive Protein: An Activator of Innate Immunity and a Modulator of Adaptive Immunity. Immunol. Res. 2004, 30, 261–278.
  109. Hage, F.G.; Szalai, A.J. C-Reactive Protein Gene Polymorphisms, C-Reactive Protein Blood Levels, and Cardiovascular Disease Risk. J. Am. Coll. Cardiol. 2007, 50, 1115–1122.
  110. Azoulay, D.; Shehadeh, M.; Chepa, S.; Shaoul, E.; Baroum, M.; Horowitz, N.A.; Kaykov, E. Recovery from SARS-CoV-2 infection is associated with serum BDNF restoration. J. Infect. 2020, 81, e79–e81.
  111. Bhargava, A.; Fukushima, E.A.; Levine, M.; Zhao, W.; Tanveer, F.; Szpunar, S.M.; Saravolatz, L. Predictors for Severe COVID-19 Infection. Clin. Infect. Dis. 2020, 71, 1962–1968.
  112. Liu, Q.; Dai, Y.; Feng, M.; Wang, X.; Liang, W.; Yang, F. Associations between serum amyloid A, interleukin-6, and COVID-19: A cross-sectional study. J. Clin. Lab. Anal. 2020, 34, e23527.
  113. Pincemail, J.; Cavalier, E.; Charlier, C.; Cheramy–Bien, J.-P.; Brevers, E.; Courtois, A.; Fadeur, M.; Meziane, S.; Goff, C.; Misset, B.; et al. Oxidative Stress Status in COVID-19 Patients Hospitalized in Intensive Care Unit for Severe Pneumonia. A Pilot Study. Antioxidants 2021, 10, 257.
  114. Wang, C.; Hu, S.; Wang, L.; Li, M.; Li, H. Early risk factors of the exacerbation of coronavirus disease 2019 pneumonia. J. Med. Virol. 2020, 92, 2593–2599.
  115. Song, J.-W.; Lam, S.M.; Fan, X.; Cao, W.-J.; Wang, S.-Y.; Tian, H.; Chua, G.H.; Zhang, C.; Meng, F.-P.; Xu, Z.; et al. Omics-Driven Systems Interrogation of Metabolic Dysregulation in COVID-19 Pathogenesis. Cell Metab. 2020, 32, 188–202.e5.
  116. Yuan, B.; Li, W.; Liu, H.; Cai, X.; Song, S.; Zhao, J.; Hu, X.; Li, Z.; Chen, Y.; Zhang, K.; et al. Correlation between immune response and self-reported depression during convalescence from COVID-19. Brain Behav. Immun. 2020, 88, 39–43.
  117. Guo, Q.; Zheng, Y.; Shi, J.; Wang, J.; Li, G.; Li, C.; Fromson, J.A.; Xu, Y.; Liu, X.; Xu, H.; et al. Immediate psychological distress in quarantined patients with COVID-19 and its association with peripheral inflammation: A mixed-method study. Brain Behav. Immun. 2020, 88, 17–27.
  118. Boyman, O.; Sprent, J. The role of interleukin-2 during homeostasis and activation of the immune system. Nat. Rev. Immunol. 2012, 12, 180–190.
  119. Srivastava, K.; Goyal, S.; Kodange, C.; Bhat, P.S. Immunological changes in depression. Ind. Psychiatry J. 2017, 26, 201–206.
  120. Yoon, H.-K.; Kim, Y.-K.; Lee, H.-J.; Kwon, D.-Y.; Kim, L. Role of cytokines in atypical depression. Nord. J. Psychiatry 2011, 66, 183–188.
  121. Capuron, L.; Miller, A.H. Cytokines and psychopathology: Lessons from interferon-α. Biol. Psychiatry 2004, 56, 819–824.
  122. 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.
  123. 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.
  124. Carvalho, A.F.; Solmi, M.; Sanches, M.; Machado, M.O.; Stubbs, B.; Ajnakina, O.; Sherman, C.; Sun, Y.R.; Liu, C.S.; Brunoni, A.R.; et al. Evidence-based umbrella review of 162 peripheral biomarkers for major mental disorders. Transl. Psychiatry 2020, 10, 1–13.
  125. Liu, Y.; Ho, R.C.-M.; Mak, A. Interleukin (IL)-6, tumour necrosis factor alpha (TNF-α) and soluble interleukin-2 receptors (sIL-2R) are elevated in patients with major depressive disorder: A meta-analysis and meta-regression. J. Affect. Disord. 2012, 139, 230–239.
  126. Goldsmith, D.R.; Rapaport, M.H.; Miller, B.J. A meta-analysis of blood cytokine network alterations in psychiatric patients: Comparisons between schizophrenia, bipolar disorder and depression. Mol. Psychiatry 2016, 21, 1696–1709.
  127. Qin, C.; Zhou, L.; Hu, Z.; Zhang, S.; Yang, S.; Tao, Y.; Xie, C.; Ma, K.; Shang, K.; Wang, W.; et al. Dysregulation of Immune Response in Patients with COVID-19 in Wuhan, China. SSRN Electron. J. 2020, 71, 762–768.
  128. Zhang, Y.; Wang, X.; Li, X.; Xi, D.; Mao, R.; Wu, X.; Cheng, S.; Sun, X.; Yi, C.; Ling, Z.; et al. Potential contribution of increased soluble IL-2R to lymphopenia in COVID-19 patients. Cell. Mol. Immunol. 2020, 17, 878–880.
  129. Kaya, H.; Kaji, M.; Usuda, D. Soluble interleukin-2 receptor levels on admission associated with mortality in coronavirus disease. Int. J. Infect. Dis. 2021, 105, 522–524.
  130. Hou, H.; Zhang, B.; Huang, H.; Luo, Y.; Wu, S.; Tang, G.; Liu, W.; Mao, L.; Wang, F.; Sun, Z. Using IL-2R/lymphocytes for predicting the clinical progression of patients with COVID-19. Clin. Exp. Immunol. 2020, 201, 76–84.
  131. Moshage, H.; Roelofs, H.; van Pelt, J.; Hazenberg, B.; van Leeuwen, M.; Limburg, P.; Aarden, L.; Yap, S. The effect of interleukin-1, interleukin-6 and its interrelationship on the synthesis of serum amyloid A and C-reactive protein in primary cultures of adult human hepatocytes. Biochem. Biophys. Res. Commun. 1988, 155, 112–117.
  132. Eklund, K.K.; Niemi, K.; Kovanen, P.T. Immune Functions of Serum Amyloid A. Crit. Rev. Immunol. 2012, 32, 335–348.
  133. Esen, N.; Tanga, F.Y.; DeLeo, J.A.; Kielian, T. Toll-like receptor 2 (TLR2) mediates astrocyte activation in response to the Gram-positive bacterium Staphylococcus aureus. J. Neurochem. 2003, 88, 746–758.
  134. Yu, Y.; Liu, J.; Li, S.-Q.; Peng, L.; Ye, R.D. Serum Amyloid A Differentially Activates Microglia and Astrocytes via the PI3K Pathway. J. Alzheimer’s Dis. 2013, 38, 133–144.
  135. Wang, Q.; Su, X.; Jiang, X.; Dong, X.; Fan, Y.; Zhang, J.; Yu, C.; Gao, W.; Shi, S.; Jiang, J.; et al. iTRAQ technology-based identification of human peripheral serum proteins associated with depression. Neuroscience 2016, 330, 291–325.
  136. Van Dooren, F.E.; Schram, M.; Schalkwijk, C.G.; Stehouwer, C.D.; Henry, R.M.; Dagnelie, P.C.; Schaper, N.; van der Kallen, C.J.; Koster, A.; Sep, S.J.; et al. Associations of low grade inflammation and endothelial dysfunction with depression—The Maastricht Study. Brain Behav. Immun. 2016, 56, 390–396.
  137. Silva-Costa, L.C.; Carlson, P.T.; Guest, P.C.; de Almeida, V.; Martins-De-Souza, D. Proteomic Markers for Depression. In Reviews on Biomarker Studies in Psychiatric and Neurodegenerative Disorders; Springer: Berlin/Heidelberg, Germany, 2019; Volume 1118, pp. 191–206.
  138. Pieri, M.; Ciotti, M.; Nuccetelli, M.; Perrone, M.A.; Caliò, M.T.; Lia, M.S.; Minieri, M.; Bernardini, S. Serum Amyloid A Protein as a useful biomarker to predict COVID-19 patients severity and prognosis. Int. Immunopharmacol. 2021, 95, 107512.
  139. Zinellu, A.; Paliogiannis, P.; Carru, C.; Mangoni, A.A. Serum amyloid A concentrations, COVID-19 severity and mortality: An updated systematic review and meta-analysis. Int. J. Infect. Dis. 2021, 105, 668–674.
  140. Lionetto, L.; Ulivieri, M.; Capi, M.; De Bernardini, D.; Fazio, F.; Petrucca, A.; Pomes, L.M.; De Luca, O.; Gentile, G.; Casolla, B.; et al. Increased kynurenine-to-tryptophan ratio in the serum of patients infected with SARS-CoV2: An observational cohort study. Biochim. Biophys. Acta (BBA)—Mol. Basis Dis. 2020, 1867, 166042.
  141. Danlos, F.-X.; Grajeda-Iglesias, C.; Durand, S.; Sauvat, A.; Roumier, M.; Cantin, D.; Colomba, E.; Rohmer, J.; Pommeret, F.; Baciarello, G.; et al. Metabolomic analyses of COVID-19 patients unravel stage-dependent and prognostic biomarkers. Cell Death Dis. 2021, 12, 1–11.
  142. Viana, S.D.; Nunes, S.; Reis, F. ACE2 imbalance as a key player for the poor outcomes in COVID-19 patients with age-related comorbidities—Role of gut microbiota dysbiosis. Ageing Res. Rev. 2020, 62, 101123.
  143. Zhang, H.; Li, H.-B.; Lyu, J.-R.; Lei, X.-M.; Li, W.; Wu, G.; Lyu, J.; Dai, Z.-M. Specific ACE2 expression in small intestinal enterocytes may cause gastrointestinal symptoms and injury after 2019-nCoV infection. Int. J. Infect. Dis. 2020, 96, 19–24.
  144. Bouças, A.P.; Rheinheimer, J.; Lagopoulos, J. Why Severe COVID-19 Patients Are at Greater Risk of Developing Depression: A Molecular Perspective. Neuroscience 2020.
  145. Angelucci, F.; Brenè, S.; Mathé, A.A. BDNF in schizophrenia, depression and corresponding animal models. Mol. Psychiatry 2005, 10, 345–352.
  146. Binder, D.K.; Scharfman, H.E. Mini Review. Growth Factors 2004, 22, 123–131.
  147. Bathina, S.; Das, U.N. Brain-derived neurotrophic factor and its clinical implications. Arch. Med. Sci. 2015, 6, 1164–1178.
  148. Nestler, E.J.; Barrot, M.; DiLeone, R.; Eisch, A.; Gold, S.J.; Monteggia, L.M. Neurobiology of Depression. Neuron 2002, 34, 13–25.
  149. Polyakova, M.; Stuke, K.; Schuemberg, K.; Mueller, K.; Schoenknecht, P.; Schroeter, M.L. BDNF as a biomarker for successful treatment of mood disorders: A systematic & quantitative meta-analysis. J. Affect. Disord. 2015, 174, 432–440.
  150. Gervasoni, N.; Aubry, J.-M.; Bondolfi, G.; Osiek, C.; Schwald, M.; Bertschy, G.; Karege, F. Partial Normalization of Serum Brain-Derived Neurotrophic Factor in Remitted Patients after a Major Depressive Episode. Neuropsychobiology 2005, 51, 234–238.
  151. Aydemir, O.; Deveci, A.; Taneli, F. The effect of chronic antidepressant treatment on serum brain-derived neurotrophic factor levels in depressed patients: A preliminary study. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2005, 29, 261–265.
  152. Wang, X.-L.; Iwanami, J.; Min, L.-J.; Tsukuda, K.; Nakaoka, H.; Bai, H.-Y.; Shan, B.-S.; Kan-No, H.; Kukida, M.; Chisaka, T.; et al. Deficiency of angiotensin-converting enzyme 2 causes deterioration of cognitive function. NPJ Aging Mech. Dis. 2016, 2, 16024.
  153. Verdecchia, P.; Cavallini, C.; Spanevello, A.; Angeli, F. The pivotal link between ACE2 deficiency and SARS-CoV-2 infection. Eur. J. Intern. Med. 2020, 76, 14–20.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 418
Revisions: 2 times (View History)
Update Date: 28 Jan 2022
1000/1000