Abstract

The Human Microbiome Project, launched in in 2007, and the discovery of innate lymphoid cells (ILCs) in 2008 are major advances that have begun to shed light on the pathogenesis of some disorders of uncertain etiology, including autoimmune, fibrotic, and neuropsychiatric illnesses, as well as the potential interaction of systemic infections with the gut-brain axis.

Downloads

Download data is not yet available.

“All disease begins in the gut”

Hippocrates:

The Human Microbiome Project, launched in in 2007, and the discovery of innate lymphoid cells (ILCs) in 2008 are major advances that have begun to shed light on the pathogenesis of some disorders of uncertain etiology, including autoimmune, fibrotic, and neuropsychiatric illnesses, as well as the potential interaction of systemic infections with the gut-brain axis.

The gut microbial community is immunologically “tolerated” in the gastrointestinal (GI) tract but may elicit immunogenicity and pathology upon translocation into host tissues (1) (2). Indeed, in the former case long-term, low-grade inflammation may eventually lead to disease (3), while in the latter case, numerous studies have reported the presence of intestinal microbes and/or their molecules as part of observed pathology within host tissues, such as the circulatory system and the brain (4) (5).

A better understanding of oral and gut tolerance, immune unresponsiveness to food proteins and gut microbiota, has led to the reconceptualization of some idiopathic diseases as microbial translocation disorders (MTDs) (6). Ironically, disease caused by viruses that were originally considered unrelated, such as human immunodeficiency virus (HIV) and severe acute respiratory syndrome coronaviruses (SARS and SARS-CoV-2) have contributed to a more complete understanding of the molecular underpinnings of oral/gut tolerance and immunogenicity (7)(8). As both HIV and SARS-CoV-2 viruses target ILC type 3 (ILC3), they disrupt the intestinal barrier and the immunological tolerance of oral and gut microbiome (9)(10)(11)(12). Indeed, ILC3-generated interleukin 22 (IL22) functions as a guardian of the intestinal barrier as it upregulates the antimicrobial peptides and luminal mucus, opposing microbial translocation (13)(14) (Fig. 1).

Figure 1. HIV induces apoptotic loss of ILC3, lowering IL22, the guardian of gut barrier, allowing translocation of intestinal microbes and their molecules into the systemic circulation. Activated host immunity maintains a state of low-grade inflammation that characterizes many chronic diseases of uncertain etiology.

Innate lymphoid cells:

ILCs are non-T and non-B lymphocytes, consisting of natural killer cells (NKCs), ILC-1, ILC-2, ILC-3, that reside in various tissues, including the central nervous system (CNS) and the GI tract (15)(Fig. 1). These cells play a key role in gut immunological tolerance as they maintain the barrier integrity and homeostasis, increasing antimicrobial peptides and intestinal mucus (16)(17).

Unlike the B and T cells, ILCs do not possess specific antigen receptors but express transcription factors and synthesize cytokines (18). For example, NKCs and ILC1, activated by IL12, IL15, and IL18 release interferon γ (IFN-γ), participating in antiviral defenses (Fig. 1). ILC-2 express GATA-3, are activated by IL-25 and IL33, and release IL5 and IL13; ILC3, expressing RORγt, are activated by IL23 and IL1 beta (IL-1β), while their output consists of IL22 and IL17 (Fig.2). Dysfunctional signaling in these lymphoid systems can trigger MTDs, though the full range of interactions remains to be determined.

Figure 2. Innate lymphoid cells are comprised of NKCs, ILC1, ILC2, and ILC3. They are activated by various cytokines (top), release other cytokines (bottom), and express transcription factors, including T-bet, GATA-3 and RORγt. When dysregulated, these lymphoid systems may trigger autoimmunity, fibrotic and neuropsychiatric illnesses.

In the following sections, we highlight several mechanisms that may connect ILCs with the disorders of unknown etiology, especially autoimmune, fibrotic, and neuropsychiatric illnesses

Autoimmune disorders :

Autoimmune disorders are believed to reflect an immune system dysfunction characterized by the generation of autoantibodies against self-proteins. However, improved understanding of the microbiome and ILCs has contributed to the emergence of noncanonical views in which autoantibodies can be conceptualized as conventional immunoglobulins directed at the translocated gut microbes and/or their antigens that resemble human proteins. As gut commensals express receptors similar to those of the human host, translocated microbes may elicit antibodies against these proteins, triggering pathology. For example, lupus-associated autoantigen Ro60, expressed by numerous gut commensal species, may elicit conventional immune responses when these microbes migrate into the host systemic circulation (19)(20). In another example, the gut resident Escherichia coli, expressing succinate dehydrogenase, a molecule that mimics the human mitochondrial enzyme, may elicit the formation of antimitochondrial antibodies documented in several autoimmune disorders (21)(22). Interestingly, elevated plasma succinate levels were associated with cardiovascular disease, hypertension, and diabetes type 2 (T2D), connecting these diseases of uncertain/multiple etiology to microbial translocation (23)(24). Along this line, a recent study has associated suicidal behavior in young adults with the depletion of succinate producing Alloprevotella rava, linking this condition to MTD (25). This is not the first time that suicidal behavior has been linked to potential enzymatic imbalance (26)(27), however it is one of the first linking directly to a specific species within the microbiome. Furthermore, diabetes type 1 (T1D) was associated with Citrobacter rodentium, suggesting a probable MTD etiology (28). Indeed, several studies have linked both T1D and suicidal behavior to increased levels of intestinal fatty acid binding protein (I-FABP), a serological biomarker of dysfunctional intestinal barrier, further emphasizing the likely MST etiology (29)(30). Moreover, succinate dehydrogenase was implicated in lung fibrosis, and a number of cancers, highlighting the interconnectedness of several diseases of unknown etiology (31)(32).

Neuropsychiatric disorders :

Upregulated translocation markers, I-FABP and lipopolysaccharide (LPS) were reported in several neuropsychiatric conditions, including major depressive disorder (MDD) and Alzheimer’s disease (AD), linking these pathologies to MTDs (33)(34)(35)(36). This common biological foundation is further substantiated by the higher prevalence of several neuropsychiatric conditions, such as MDD and schizophrenia, in patients with inflammatory bowel disease (IBD), a disorder marked by impaired gut barrier (37)(38)(39). Moreover, CNS-resident ILCs have been implicated in MDD, neurodegeneration, multiple sclerosis (MS), and suicidal behavior, connecting these pathologies to MTDs (40)(41)(42)(43). Indeed, dysfunctional ILC2 and IL15 and IL13 mark autoimmune disorders as well as schizophrenia and AD, linking these neuropsychiatric conditions to autoantibodies (44)(45)(46)(47)(48). This is significant as autoantibodies against neuronal adhesion molecule (NCAM1) were recently detected in patients with Guillan-Barre Syndrome (49) and schizophrenia, emphasizing that inflammation in these conditions may be autoimmune in nature (50). As NCAM1 has been associated with several viruses, including COVID-19, rabies, and Zika, non-familial schizophrenia may be the result of prenatal exposure to those and similar pathogens as demonstrated in the aftermath of the 1957 influenza A2 epidemic (51)(52)(53)(54). In addition, as IL-13 attachment to its α1 receptor (IL13R α1) can trigger loss of dopaminergic neurons in substantia nigra, ILC3 may be implicated in Parkinson’s disease (55)(56). Moreover, IL13 was reported to differentiate between MDD patients with and without suicidal intent, suggesting that for a portion of neuropsychiatric patients this cytokine could become a biomarker of suicidality (57)(58).

Various gut microbes produce molecules mimicking gamma-aminobutyric acid (GABA) and glutamic acid decarboxylase (GAD or GAD65), suggesting that microbiota translocation may elicit autoantibodies directed at these proteins (59). Indeed, anti-GAD antibodies were demonstrated in schizophrenia, bipolar disorder, T1D, and autoimmune thyroiditis, further emphasizing the intertwined nature of the idiopathic disorders (60). Interestingly, the protozoan Toxoplasma gondii (T. gondii), previously associated with schizophrenia, utilizes GABA as a carbon source, depleting this neurotransmitter, likely contributing to neuropathology (61)(62). Moreover, T. gondii was found to induce immunosuppression via IL-10, NKCs and ILC1, implicating both immunodeficiency and autoimmunity in T. gondii psychopathology (63)(64). This is significant as several studies have demonstrated a high rate of latent T. gondii infection among COVID-19 patients, indicating a possible opportunistic symbiosis for disruption of host immunity (65).

Human fibrotic diseases

Fibrotic diseases, marked by the accumulation of excessive extracellular matrix proteins (EMPs), encompass both generalized and organ-specific disorders, including systemic sclerosis, pulmonary, cardiac, intestinal, liver, and kidney fibrosis (66). Transforming growth factor-β (TGF-β), upregulated in fibrotic illness, is a key regulator of ILCs, linking excessive fibrosis to MTDs (67)(68)(69)(70)(71). Indeed, fibrosis has been associated with autoantibodies and the breakdown of gut immunological tolerance. For example, microbiota-generated curli amyloid fibrils were reported to disrupt ILC3, IL17, and IL22, promoting both autoimmunity and intestinal fibrosis (72)(73)(74)(75)(76). Furthermore, fibroblast growth factors play a key role in GI barrier integrity, while loss of fibroblast growth factor 9 (FGF9) was associated with pulmonary fibrosis, further linking this pathology to MTDs (77)(78)(79). This is significant as FGF9 possesses antiviral properties, probably regulating the gut virome, an intestinal viral community associated with various pathologies, including fibrosis, autoimmunity, and neuropsychiatric illness (80)(81)(82)(83)(84). Indeed, FGF9 has been implicated in MS, seizure disorder, and schizophrenia, emphasizing once more the interconnectedness of these poorly understood disorders (85)(86)(87).

While this overview provides a first indication of the potential links between MTDs, auto-immune and neuro-psychiatric disorders, it should be noted that a number of these investigations remain in their early stage. Additionally, these conditions have been presented here as being well-distinct, though in practice there are observed areas of etiological overlap. Therefore, caution would need to be exercised in inferring causation from apparent correlation. While there is no doubt that the discovery of ILCs and the improved understanding of the gut-brain axis will contribute to the in-depth knowledge of conditions and pathologies of as yet unknown etiology (and eventually lead to novel treatments), there exist profound gaps in the current scientific knowledge that require to be addressed by further molecular and population-based studies on the subject.

Conclusions

The discovery of ILCs contributed to a better understanding of microbial translocation outside the GI tract. The host-microbiota interaction may elucidate the etiology of some poorly understood disorders, including autoimmunity, fibrotic illness, and neuropsychiatric conditions. Restoring ILCs homeostasis and intestinal barrier integrity to oppose microbial translocation will, eventually lead to the development of novel therapeutic strategies for chronic illnesses.

Disclaimer

Where authors are identified as personnel of the International Agency for Research on Cancer/WHO, the authors alone are responsible for the views expressed in this article and they do not necessarily represent the decisions, policy or views of the International Agency for Research on Cancer/WHO.

References

  1. Ha CWY, Martin A, Sepich-Poore GD, Shi B, Wang Y, Gouin K, Humphrey G, et al. Translocation of Viable Gut Microbiota to Mesenteric Adipose Drives Formation of Creeping Fat in Humans. Cell. 2020 Oct 29;183(3):666-683.e17. Doi: 10.1016/j.cell.2020.09.009.
  2. Vaishnavi C. Translocation of gut flora and its role in sepsis. Indian J Med Microbiol. 2013 Oct-Dec;31(4):334-42. Doi: 10.4103/0255-0857.118870. PMID: 24064638.
  3. van den Munckhof ICL, Kurilshikov A, Ter Horst R, Riksen NP, Joosten LAB, Zhernakova A, Fu J, Keating ST, Netea MG, de Graaf J, Rutten JHW. Role of gut microbiota in chronic low-grade inflammation as potential driver for atherosclerotic cardiovascular disease: a systematic review of human studies. Obes Rev. 2018 Dec;19(12):1719-1734. Doi: 10.1111/obr.12750.
  4. D'Aquila P, Giacconi R, Malavolta M, Piacenza F, Bürkle A, Villanueva MM, et al.. Microbiome in Blood Samples From the General Population Recruited in the MARK-AGE Project: A Pilot Study. Front Microbiol.
  5. Zhao Y, Cong L, Lukiw WJ. Lipopolysaccharide (LPS) Accumulates in Neocortical Neurons of Alzheimer's disease (AD) Brain and Impairs Transcription in Human Neuronal-Glial Primary Co-cultures. Front Aging Neurosci. 2017 Dec 12;9:407. Doi: 10.3389/fnagi.2017.00407.
  6. Tordesillas L, Berin MC. Mechanisms of Oral Tolerance. Clin Rev Allergy Immunol. 2018 Oct;55(2):107-117. Doi: 10.1007/s12016-018-8680-5.
  7. Oliva A, Miele MC, Di Timoteo F, De Angelis M, Mauro V, Aronica R, et al. Persistent Systemic Microbial Translocation and Intestinal Damage During Coronavirus Disease-19. Front Immunol. 2021 Jul 14;12:708149. Doi: 10.3389/fimmu.2021.708149.
  8. Klatt NR, Funderburg NT, Brenchley JM. Microbial translocation, immune activation, and HIV disease. Trends Microbiol. 2013 Jan;21(1):6-13. Doi: 10.1016/j.tim.2012.09.001.
  9. Silverstein NJ, Wang Y, Manickas-Hill Z, Carbone C, Dauphin A, Boribong BP, et al. Innate lymphoid cells and COVID-19 severity in SARS-CoV-2 infection. Elife. 2022 Mar 11;11:e74681. doi: 10.7554/eLife.74681
  10. Panda SK, Colonna M. Innate Lymphoid Cells in Mucosal Immunity. Front Immunol. 2019 May 7;10:861. Doi: 10.3389/fimmu.2019.00861.
  11. Guo X, Fu YX. The tragic fate of group 3 innate lymphoid cells during HIV-1 infection. J Clin Invest. 2015 Sep;125(9):3430-2. Doi: 10.1172/JCI83823. Epub 2015 Aug 24. Erratum in: J Clin Invest. 2015 Oct 1;125(10):3992.
  12. Kløverpris HN, Kazer SW, Mjösberg J, Mabuka JM, Wellmann A, Ndhlovu Z, et al. Innate Lymphoid Cells Are Depleted Irreversibly during Acute HIV-1 Infection in the Absence of Viral Suppression. Immunity. 2016 Feb 16;44(2):391-405. Doi: 10.1016/j.immuni.2016.01.006.
  13. Wei HX, Wang B, Li B. IL-10 and IL-22 in Mucosal Immunity: Driving Protection and Pathology. Front Immunol. 2020 Jun 26;11:1315. Doi: 10.3389/fimmu.2020.01315.
  14. Keir M, Yi Y, Lu T, Ghilardi N. The role of IL-22 in intestinal health and disease. J Exp Med. 2020;217(3):e20192195.
  15. Vivier E, Artis D, Colonna M, Diefenbach A, Di Santo JP, Eberl G, et al. Innate Lymphoid Cells: 10 Years On. Cell. 2018 Aug 23;174(5):1054-1066. Doi: 10.1016/j.cell.2018.07.017.
  16. Artis, D., Spits, H. The biology of innate lymphoid cells. Nature 517, 293–301 (2015). https://doi.org/10.1038/nature14189
  17. Fan H, Wang A, Wang Y, Sun Y, Han J, Chen W, Wang S, Wu Y, Lu Y. Innate Lymphoid Cells: Regulators of Gut Barrier Function and Immune Homeostasis. J Immunol Res. 2019 Dec 20;2019:2525984. Doi: 10.1155/2019/2525984.
  18. Crinier A, Viant C, Girard-Madoux M, Vivier É. Les cellules lymphoïdes innées [Innate lymphoid cells]. Med Sci (Paris). 2017 May;33(5):534-542. French. Doi: 10.1051/medsci/20173305018.
  19. Greiling TM, Dehner C, Chen X, Hughes K, Iñiguez AJ, Boccitto M, Ruiz DZ, et al. Commensal orthologs of the human autoantigen Ro60 as triggers of autoimmunity in lupus. Sci Transl Med. 2018 Mar 28;10(434):eaan2306. doi: 10.1126/scitranslmed.aan2306.
  20. Boccitto M, Wolin SL. Ro60 and Y RNAs: structure, functions, and roles in autoimmunity. Crit Rev Biochem Mol Biol. 2019 Apr;54(2):133-152. Doi: 10.1080/10409238.2019.1608902.
  21. Kemp EH, Ridgway JN, Smith KA, Watson PF, Weetman AP. Autoantibodies to the flavoprotein subunit of succinate dehydrogenase: analysis of specificity in autoimmune thyroid disease. Clin Endocrinol (Oxf). 2000 Sep;53(3):291-9. Doi: 10.1046/j.1365-2265.2000.01072.x. PMID: 10971445.
  22. Tran QM, Rothery RA, Maklashina E, Cecchini G, Weiner JH. Escherichia coli succinate dehydrogenase variant lacking the heme b. Proc Natl Acad Sci U S A. 2007 Nov 13;104(46):18007-12. doi: 10.1073/pnas.0707732104.
  23. Osuna-Prieto FJ, Martinez-Tellez B, Ortiz-Alvarez L, Di X, Jurado-Fasoli L, Xu H, Ceperuelo-Mallafré V, et al. Elevated plasma succinate levels are linked to higher cardiovascular disease risk factors in young adults. Cardiovasc Diabetol. 2021 Jul 27;20(1):151. Doi: 10.1186/s12933-021-01333-3.
  24. Serena C, Ceperuelo-Mallafré V, Keiran N, Queipo-Ortuño MI, Bernal R, Gomez-Huelgas R, et al. Elevated circulating levels of succinate in human obesity are linked to specific gut microbiota. ISME J. 2018 Jun;12(7):1642-1657. Doi: 10.1038/s41396-018-0068-2.
  25. Ahrens AP, Sanchez-Padilla DE, Drew JC, Oli MW, Roesch LFW, Triplett EW. Saliva microbiome, dietary, and genetic markers are associated with suicidal ideation in university students. Sci Rep. 2022 Aug 22;12(1):14306. Doi: 10.1038/s41598-022-18020-2.
  26. Brundin, L., Bryleva, E. & Thirtamara Rajamani, K. Role of Inflammation in Suicide: From Mechanisms to Treatment. Neuropsychopharmacol 42, 271–283 (2017). https://doi.org/10.1038/npp.2016.116
  27. Keshri N, Nandeesha H, Kattimani S. Elevated interleukin-17 and reduced testosterone in bipolar disorder. Relation with suicidal behaviour. Asian J Psychiatr. 2018 Aug;36:66-68. doi: 10.1016/j.ajp.2018.06.011
  28. Pöysti, S., Toivonen, R., Takeda, A. et al. Infection with the enteric pathogen C. rodentium promotes islet-specific autoimmunity by activating a lymphatic route from the gut to pancreatic lymph node. Mucosal Immunol 15, 471–479 (2022). https://doi.org/10.1038/s41385-022-00490-2
  29. Ohlsson L, Gustafsson A, Lavant E, Suneson K, Brundin L, Westrin Å, et al. Leaky gut biomarkers in depression and suicidal behavior. Acta Psychiatr Scand. 2019 Feb;139(2):185-193. Doi: 10.1111/acps.12978. Epub 2018 Nov 1. Erratum in: Acta Psychiatr Scand. 2020 Nov;142(5):423. PMID: 30347427; PMCID: PMC6587489.
  30. Ochocińska A, Wysocka-Mincewicz M, Groszek A, Rybak A, Konopka E, Bierła JB. Could I-FABP Be an Early Marker of Celiac Disease in Children with Type 1 Diabetes? Retrospective Study from the Tertiary Reference Centre. Nutrients. 2022 Jan 18;14(3):414. Doi: 10.3390/nu14030414.
  31. Wang Z, Chen L, Huang Y, Luo M, Wang H, Jiang Z, et al. Pharmaceutical targeting of succinate dehydrogenase in fibroblasts controls bleomycin-induced lung fibrosis. Redox Biol. 2021 Oct;46:102082. Doi: 10.1016/j.redox.2021.102082.
  32. Zhao T, Mu X, You Q. Succinate: An initiator in tumorigenesis and progression. Oncotarget. 2017 May 10;8(32):53819-53828. Doi: 10.18632/oncotarget.17734.
  33. Arnone D. Increased levels of intestinal-type fatty acid-binding protein (I-FABP) in mood disorders. Psychol Med. 2022 Jul 21:1-2. doi: 10.1017/S0033291722001970
  34. Zhan X, Stamova B, Sharp FR. Lipopolysaccharide Associates with Amyloid Plaques, Neurons and Oligodendrocytes in Alzheimer's disease Brain: A Review. Front Aging Neurosci. 2018 Feb 22;10:42. Doi: 10.3389/fnagi.2018.00042.
  35. Meier-Stephenson FS, Meier-Stephenson VC, Carter MD, Meek AR, Wang Y, Pan L, Chen Q, et al. Alzheimer's disease as an autoimmune disorder of innate immunity endogenously modulated by tryptophan metabolites. Alzheimers Dement (N Y). 2022 Apr 6;8(1):e12283. doi: 10.1002/trc2.12283
  36. Euesden J, Danese A, Lewis CM, Maughan B. A bidirectional relationship between depression and the autoimmune disorders - New perspectives from the National Child Development Study. PLoS One. 2017 Mar 6;12(3):e0173015. Doi: 10.1371/journal.pone.0173015.
  37. Iordache MM, Tocia C, Aschie M, Dumitru A, Manea M, Cozaru GC, et al. Intestinal Permeability and Depression in Patients with Inflammatory Bowel Disease. J Clin Med. 2022 Aug 30;11(17):5121. Doi: 10.3390/jcm11175121.
  38. Ishida I, Ogura J, Aizawa E, Ota M, Hidese S, Yomogida Y, Matsuo J, Yoshida S, Kunugi H. Gut permeability and its clinical relevance in schizophrenia. Neuropsychopharmacol Rep. 2022 Mar;42(1):70-76. Doi: 10.1002/npr2.12227.
  39. Sung KY, Zhang B, Wang HE, Bai YM, Tsai SJ, Su TP, Chen TJ, Hou MC, Lu CL, Wang YP, Chen MH. Schizophrenia and risk of new-onset inflammatory bowel disease: a nationwide longitudinal study. Aliment Pharmacol Ther. 2022 May;55(9):1192-1201. Doi: 10.1111/apt.16856.
  40. Wang S, van de Pavert SA. Innate Lymphoid Cells in the Central Nervous System. Front Immunol. 2022 Feb 3;13:837250. Doi: 10.3389/fimmu.2022.837250.
  41. Barichello T. The role of innate lymphoid cells (ILCs) in mental health. Discov Ment Health. 2022;2(1):2. Doi: 10.1007/s44192-022-00006-1. Epub 2022 Feb 7. PMID: 35224555; PMCID: PMC8855986.
  42. Madeshiya AK, Pillai A. Innate lymphoid cells in depression: Current status and perspectives, Biomarkers in Neuropsychiatry, Volume 7, 2022, https://doi.org/10.1016/j.bionps.2022.100055.
  43. Yeung SS, Ho YS, Chang RC. The role of meningeal populations of type II innate lymphoid cells in modulating neuroinflammation in neurodegenerative diseases. Exp Mol Med. 2021;53(9):1251–67.
  44. Yeh TC, Chu HT, Tsai CK, Chang HA, Yang FC, Huang SY, Liang CS. Distinct Inflammation Biomarkers in Healthy Individuals and Patients with Schizophrenia: A Reliability Testing of Multiplex Cytokine Immunoassay by Bland-Altman Analysis. Psychiatry Investig. 2019 Aug;16(8):607-614. Doi: 10.30773/pi.2019.04.14.
  45. Fung ITH, Zhang Y, Shin DS, Sankar P, Sun X, D'Souza SS, Song R, Kuentzel ML, Chittur SV, Zuloaga KL, Yang Q. Group 2 innate lymphoid cells are numerically and functionally deficient in the triple transgenic mouse model of Alzheimer's disease. J Neuroinflammation. 2021 Jul 6;18(1):152. Doi: 10.1186/s12974-021-02202-2.
  46. 42.Jeong JY, Chung YC, Jin BK. Interleukin-4 and Interleukin-13 Exacerbate Neurotoxicity of Prothrombin Kringle-2 in Cortex In Vivo via Oxidative Stress. Int J Mol Sci. 2019 Apr 19;20(8):1927. Doi: 10.3390/ijms20081927.
  47. Momtazmanesh S, Zare-Shahabadi A, Rezaei N. Cytokine Alterations in Schizophrenia: An Updated Review. Front Psychiatry. 2019 Dec 6;10:892. Doi: 10.3389/fpsyt.2019.00892.
  48. Laskaris L, Mancuso S, Shannon Weickert C, Zalesky A, Chana G, et al. Brain morphology is differentially impacted by peripheral cytokines in schizophrenia-spectrum disorder. Brain Behav Immun. 2021 Jul;95:299-309. doi: 10.1016/j.bbi.2021.04.002
  49. Shiwaku H, Katayama S, Kondo K, Nakano Y, Tanaka H, Yoshioka Y, et al. Autoantibodies against NCAM1 from patients with schizophrenia cause schizophrenia-related behavior and changes in synapses in mice. Cell Rep Med. 2022 Apr 19;3(4):100597. Doi: 10.1016/j.xcrm.2022.100597.
  50. Laudanski, K.; Yakhkind, A.; Restrepo, M.; Draham, L.; Lang, A.E. Guillain–Barré Syndrome in COVID-19—The Potential Role of NCAM-1 and Immunotherapy. BioMed 2021, 1, 80-92. https://doi.org/10.3390/biomed1010006
  51. Morsy S. NCAM protein and SARS-COV-2 surface proteins: In-silico hypothetical evidence for the immunopathogenesis of Guillain-Barré syndrome. Med Hypotheses. 2020 Dec;145:110342. Doi: 10.1016/j.mehy.2020.110342.
  52. Srivastava M, Zhang Y, Chen J, Sirohi D, Miller A, Zhang Y, Chen Z, Lu H, Xu J, Kuhn RJ, Andy Tao W. Chemical proteomics tracks virus entry and uncovers NCAM1 as Zika virus receptor. Nat Commun. 2020 Aug 4;11(1):3896. Doi: 10.1038/s41467-020-17638-y.
  53. Mednick SA, Machon RA, Huttunen MO, Bonett D. Adult schizophrenia following prenatal exposure to an influenza epidemic. Arch Gen Psychiatry. 1988 Feb;45(2):189-92. Doi: 10.1001/archpsyc.1988.01800260109013.
  54. Morrison BE, Marcondes MC, Nomura DK, Sanchez-Alavez M, Sanchez-Gonzalez A, Saar I, et al. IL-13Rα1 expression in dopaminergic neurons contributes to their oxidative stress-mediated loss following chronic peripheral treatment with lipopolysaccharide. J Immunol. 2012 Dec 15;189(12):5498-502. Doi: 10.4049/jimmunol.1102150.
  55. Schnell, M., McGettigan, J., Wirblich, C. et al. The cell biology of rabies virus: using stealth to reach the brain. Nat Rev Microbiol 8, 51–61 (2010). https://doi.org/10.1038/nrmicro2260
  56. Mori S, Sugama S, Nguyen W, Michel T, Sanna MG, Sanchez-Alavez M, et al. Lack of interleukin-13 receptor α1 delays the loss of dopaminergic neurons during chronic stress. J Neuroinflammation. 2017 Apr 21;14(1):88. Doi: 10.1186/s12974-017-0862-1.
  57. Ganança L, Oquendo MA, Tyrka AR, Cisneros-Trujillo S, Mann JJ, Sublette ME. The role of cytokines in the pathophysiology of suicidal behavior. Psychoneuroendocrinology. 2016 Jan;63:296-310. Doi: 10.1016/j.psyneuen.2015.10.008.
  58. Benedetta Vai, Mario Gennaro Mazza, Silvia Cazzetta, Federico Calesella, Veronica Aggio, Cristina Lorenzi, et al. Higher Interleukin 13 differentiates patients with a positive history of suicide attempts in major depressive disorder. Journal of Affective Disorders Reports, Volume 6, 2021, 100254, ISSN 2666-9153, https://doi.org/10.1016/j.jadr.2021.100254.
  59. Bedi S, Richardson TM, Jia B, Saab H, Brinkman FSL, Westley M. Similarities between bacterial GAD and human GAD65: Implications in gut mediated autoimmune type 1 diabetes. PLoS One. 2022 Feb 23;17(2):e0261103. Doi: 10.1371/journal.pone.0261103.
  60. Hansen N, Bartels C, Teegen B, Wiltfang J, Malchow B. Catatonic Schizophrenia Associated With Cerebrospinal GAD65 Autoantibodies: Case Report and Literature Review. Front Immunol. 2022 Feb 9;13:829058. Doi: 10.3389/fimmu.2022.829058.
  61. Kawasaki E, Takino H, Yano M, Uotani S, Matsumoto K, Takao Y, Yamaguchi Y, Akazawa S, Nagataki S. Autoantibodies to glutamic acid decarboxylase in patients with IDDM and autoimmune thyroid disease. Diabetes. 1994 Jan;43(1):80-6. Doi: 10.2337/diab.43.1.80.
  62. Brooks JM, Carrillo GL, Su J, Lindsay DS, Fox MA, Blader IJ. Toxoplasma gondii Infections Alter GABAergic Synapses and Signaling in the Central Nervous System. mBio. 2015 Oct 27;6(6):e01428-15. Doi: 10.1128/mBio.01428-15.
  63. Carter CJ. Schizophrenia susceptibility genes directly implicated in the life cycles of pathogens: cytomegalovirus, influenza, herpes simplex, rubella, and Toxoplasma gondii. Schizophr Bull. 2009 Nov;35(6):1163-82. Doi: 10.1093/schbul/sbn054.
  64. Vienne M, Etiennot M, Escalière B, Galluso J, Spinelli L, Guia S, et al. Type 1 Innate Lymphoid Cells Limit the Antitumoral Immune Response. Front Immunol. 2021 Nov 16;12:768989. doi: 10.3389/fimmu.2021.768989.
  65. Khan IA, Matsuura T, Kasper LH. IL-10 mediates immunosuppression following primary infection with Toxoplasma gondii in mice. Parasite Immunol. 1995 Apr;17(4):185-95. Doi: 10.1111/j.1365-3024.1995.tb00888.x. PMID: 7624159.
  66. Montazeri, M., Nakhaei, M., Fakhar, M. et al. Exploring the Association Between Latent Toxoplasma gondii Infection and COVID-19 in Hospitalized Patients: First Registry-Based Study. Acta Parasit. 67, 1172–1179 (2022). https://doi.org/10.1007/s11686-022-00559-9
  67. Wynn, T., Ramalingam, T. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat Med 18, 1028–1040 (2012). https://doi.org/10.1038/nm.2807
  68. Bottino C, Walzer T, Santoni A, Castriconi R. Editorial: TGF-β as a Key Regulator of NK and ILCs Development and Functions. Front Immunol. 2021 Jan 19;11:631712. doi: 10.3389/fimmu.2020.631712
  69. Frangogiannis N. Transforming growth factor-β in tissue fibrosis. J Exp Med. 2020 Feb 13;217(3):e20190103. doi: 10.1084/jem.20190103. PMID: 32997468; PMCID: PMC7062524.
  70. Horsburgh S, Todryk S, Ramming A, Distler JHW, O'Reilly S. Innate lymphoid cells and fibrotic regulation. Immunol Lett. 2018 Mar;195:38-44. Doi: 10.1016/j.imlet.2017.08.022. Epub 2017 Aug 24. PMID: 28844771.
  71. Majumder S, Amatya N, Revu S, Jawale CV, Wu D, Rittenhouse N, Menk A, et al. IL-17 metabolically reprograms activated fibroblastic reticular cells for proliferation and survival. Nat Immunol. 2019 May;20(5):534-545. Doi: 10.1038/s41590-019-0367-4.
  72. Forkel M, Berglin L, Kekäläinen E, Carlsson A, Svedin E, Michaëlsson J, Nagasawa M, Erjefält JS, Mori M, Flodström-Tullberg M, Bergquist A, Ljunggren HG, Westgren M, Lindforss U, Friberg D, Jorns C, Ellis E, Björkström NK, Mjösberg J. Composition and functionality of the intrahepatic innate lymphoid cell-compartment in human nonfibrotic and fibrotic livers. Eur J Immunol. 2017 Aug;47(8):1280-1294. Doi: 10.1002/eji.201646890.
  73. Nicastro LK, de Anda J, Jain N, Grando KCM, Miller AL, Bessho S, Gallucci S, Wong GCL, Tükel Ç. Assembly of ordered DNA-curli fibril complexes during Salmonella biofilm formation correlates with strengths of the type I interferon and autoimmune responses. PLoS Pathog. 2022 Aug 16;18(8):e1010742. doi: 10.1371/journal.ppat.1010742
  74. Ray S, De Salvo C, Pizarro TT. Central role of IL-17/Th17 immune responses and the gut microbiota in the pathogenesis of intestinal fibrosis. Curr Opin Gastroenterol. 2014 Nov;30(6):531-8. Doi: 10.1097/MOG.0000000000000119.
  75. Hoyne GF, Elliott H, Mutsaers SE, Prêle CM. Idiopathic pulmonary fibrosis and a role for autoimmunity. Immunol Cell Biol. 2017 Aug;95(7):577-583. Doi: 10.1038/icb.2017.22.
  76. Nishimori JH, Newman TN, Oppong GO, Rapsinski GJ, Yen JH, Biesecker SG, et al. Microbial amyloids induce interleukin 17A (IL-17A) and IL-22 responses via Toll-like receptor 2 activation in the intestinal mucosa. Infect Immun. 2012 Dec;80(12):4398-408. Doi: 10.1128/IAI.00911-12.
  77. Joannes A, Brayer S, Besnard V, Marchal-Sommé J, Jaillet M, Mordant P, Mal H, Borie R, Crestani B, Mailleux AA. FGF9 and FGF18 in idiopathic pulmonary fibrosis promote survival and migration and inhibit myofibroblast differentiation of human lung fibroblasts in vitro. Am J Physiol Lung Cell Mol Physiol. 2016 Apr 1;310(7):L615-29. Doi: 10.1152/ajplung.00185.2015.
  78. Coffey E, Newman DR, Sannes PL. Expression of fibroblast growth factor 9 in normal human lung and idiopathic pulmonary fibrosis. J Histochem Cytochem. 2013 Sep;61(9):671-9. Doi: 10.1369/0022155413497366.
  79. Danopoulos S, Schlieve CR, Grikscheit TC, Al Alam D. Fibroblast Growth Factors in the Gastrointestinal Tract: Twists and Turns. Dev Dyn. 2017 Apr;246(4):344-352. Doi: 10.1002/dvdy.24491.
  80. Zhao G, Vatanen T, Droit L, Park A, Kostic AD, Poon TW, et al. Intestinal virome changes precede autoimmunity in type I diabetes-susceptible children. Proc Natl Acad Sci U S A. 2017 Jul 25;114(30):E6166-E6175. Doi: 10.1073/pnas.1706359114. Epub 2017 Jul 10. Erratum in: Proc Natl Acad Sci U S A. 2018 Nov 27;115(48):E11426. PMID: 28696303; PMCID: PMC5544325.
  81. Yolken RH, Kinnunen PM, Vapalahti O, Dickerson F, Suvisaari J, Chen O, Sabunciyan S. Studying the virome in psychiatric disease. Schizophr Res. 2021 Aug;234:78-86. Doi: 10.1016/j.schres.2021.04.006.
  82. Ntolios P, Tzilas V, Bouros E, Avdoula E, Karakasiliotis I, Bouros D, Steiropoulos P. The Role of Microbiome and Virome in Idiopathic Pulmonary Fibrosis. Biomedicines. 2021 Apr 20;9(4):442. Doi: 10.3390/biomedicines9040442. PMID: 33924195
  83. Neurath MF, Überla K, Ng SC. Gut as viral reservoir: lessons from gut viromes, HIV and COVID-19. Gut. 2021 Sep;70(9):1605-1608. doi: 10.1136/gutjnl-2021-324622.
  84. Hiller BE, Yin Y, Perng YC, de Araujo Castro Í, Fox LE, Locke MC, et al. Fibroblast growth factor-9 expression in airway epithelial cells amplifies the type I interferon response and alters influenza A virus pathogenesis. PLoS Pathog. 2022 Jun 8;18(6):e1010228. Doi: 10.1371/journal.ppat.1010228.
  85. Clottu AS, Humbel M, Fluder N, Karampetsou MP, Comte D. Innate Lymphoid Cells in Autoimmune Diseases. Front Immunol. 2022 Jan 7;12:789788. Doi: 10.3389/fimmu.2021.789788.
  86. Guo M, Cui C, Song X, Jia L, Li D, Wang X, Dong H, Ma Y, Liu Y, Cui Z, Yi L, Li Z, Bi Y, Li Y, Liu Y, Duan W, Li C. Deletion of FGF9 in GABAergic neurons causes epilepsy. Cell Death Dis. 2021 Feb 19;12(2):196. doi: 10.1038/s41419-021-03478-1.
  87. Lindner M, Thümmler K, Arthur A, Brunner S, Elliott C, McElroy D, Mohan H, et al. Fibroblast growth factor signalling in multiple sclerosis: inhibition of myelination and induction of pro-inflammatory environment by FGF9. Brain. 2015 Jul;138(Pt 7):1875-93. Doi: 10.1093/brain/awv102.
  88. Li XL, Yu Y, Hu Y, Wu HT, Li XS, Chen GY, Cheng Y. Fibroblast Growth Factor 9 as a Potential Biomarker for Schizophrenia. Front Psychiatry. 2022 Apr 25;13:788677. Doi: 10.3389/fpsyt.2022.788677.

Figure 3.

Open Access This article is licensed under a Creative Commons Attribution 4.0 International

License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third-party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit https://creativecommons.org/licenses/by/4.0/.

© The Author(s) 2023