6
views
0
recommends
+1 Recommend
3 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found

      Will the COVID‐19 pandemic trigger future occurrence of autoimmunity like Sjögren’s syndrome?

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) is a new RNA virus that is homologous with the previously known SARS‐CoV‐2 and Middle East respiratory syndrome (MERS) coronaviruses. SARS‐CoV‐2 binds to various cell types, including lung epithelial cells, via the angiotensin‐converting enzyme (ACE)‐2 molecules expressed on the cell surface. After binding, viral RNA enters the cytosol of the infected cells and stimulates intracellular signals to activate intranuclear gene transcriptions of multiple pro‐inflammatory cytokines, including tumor necrosis factor alpha, interleukin (IL)‐6, and IL‐1, causing substantial inflammatory reactions. 1 , 2 In susceptible patients, the complex inflammatory responses, the so‐called cytokine storm, would lead to severe acute respiratory distress syndrome. 2 SARS‐CoV‐2 infection triggers autoimmunity aside from inflammation. The clinical features of the coronavirus disease 2019 (COVID‐19) may include ground‐glass opacity on chest radiography, coagulopathy, and other hematological abnormalities that resemble those found in autoimmune diseases. 3 , 4 Autoantibodies, including anti‐nuclear, anti‐Sjögren’s syndrome (SS)‐A/Ro, anti‐neutrophilic cytoplasmic, anti‐cyclic peptide containing citrulline, and anti‐interferon were detected in COVID‐19 patients. 5 , 6 , 7 As for T cell immunity, long‐term antigenic stimulation by SARS‐CoV2 would induce T cell activation and exhaustion in both CD4+ and CD8+ T cells. 8 , 9 Furthermore, a T‐helper 17 (Th17) phenotype has been shown in COVID‐19, as the T cells of patients with COVID‐19 produce more IL‐17 than those of controls. 9 T cell exhaustion and Th17 phenotype are characteristics found in autoimmune rheumatic diseases, such as in systemic lupus erythematosus (SLE). 10 In fact, a considerable number of COVID‐19 patients were diagnosed as having autoimmune rheumatic diseases as they fulfilled the respective criteria. 4 , 11 , 12 In general, infection, either viral or bacterial, is recognized as the most critical factor to accelerate the incidence and/or disease activity of autoimmune diseases. For example, parvovirus, hepatitis virus, and Epstein‐Barr virus (EBV) have been significant candidates as pathogens in autoimmune diseases such as SLE and SS. 13 , 14 The precise mechanisms by which infection would accelerates autoimmunity are unclear; however, it may be plausible that infection evokes antiviral or antibacterial immune responses and activates inflammatory cytokines, which lead to a dysregulation of innate and adaptive immunity. Pusch et al 15 described that viruses induce “heterologous immunity” to the infected host, altering adaptive immune responses to viral and self‐antigens. Moreover, EBV evokes B lymphocyte activation associated with overproduction of autoantibodies. 16 A “molecular mimicry” may exist between the viral antigen and autoantigen(s). 17 , 18 Coxsackie virus, for example, has sequence homologies between the viral protein and an autoantigen, Ro60 peptide, which is often found in SS and other autoimmune diseases. 12 In addition, infections themselves, or the administration of anti‐pathogenic agents, may alter the host's intestinal homeostasis, causing dysbiosis in the gut flora. Recent investigations suggest that the alteration of gut homeostasis, or dysbiosis, is crucial in the induction of autoimmune diseases. 19 For instance, the potential role of a species of Gram negative bacteria in oral and gut microbiota, that is, Prevotella spp., in the pathogenesis of rheumatoid arthritis is attracting widespread interest. 19 , 20 As for SS, researchers have suggested an association between gut dysbiosis and the severity of dry eye. 21 Of note, in 2011, Shoenfeld et al 22 identified the concept of ASIA syndrome (autoimmune/inflammatory syndrome induced by adjuvants). The proposed syndrome encompasses a broad spectrum of autoimmunity that is induced after exposure to external factors such as infections. 22 Considering the time gap between exposure to an infection and autoimmune disease diagnosis, the authors hypothesized that "the non‐antigenic activation” of immunity might determine the degree of autoimmune reaction after the infection. In the proposed entities for the diagnosis of “ASIA,” “exposure to external stimuli” is one of the major criteria. According to the concept, SS is a typical example of “ASIA”. 23 Considering these findings, the current pandemic of COVID‐19 would evoke the occurrence of autoimmune rheumatic diseases such as SLE or SS, 24 which may constitute the ASIA syndrome. 12 To support this notion, it was observed that a significant number of patients with COVID‐19 show clinical and laboratory findings similar to those in autoimmune rheumatic diseases. 24 The sequence homology between SARS‐CoV‐2 spike and nuclear proteins and human peptides has been demonstrated. 17 , 18 Moreover, altered gut dysbiosis after SARS‐CoV‐2 infection has been reported, 25 suggesting its implication in dysregulated immunity in COVID‐19. Regarding other coronaviruses, only few reports have described the occurrence of autoimmune diseases after SARS‐CoV or MERS‐CoV infection, owing to the relatively small‐scale pandemic compared with the current COVID‐19 pandemic. However, MERS‐CoV was reported to induce pro‐inflammatory reactions and Th17 cytokine profile, 26 suggesting a possibility that it triggers autoimmunity. Brito‐Zerón et al 27 reported that patients who developed primary SS after contracting SARS‐COV‐2 infection manifested clinical presentation similar to those in the general population. Nevertheless, Fernandez‐Gutierrez et al 28 demonstrated a slight but significant difference in the crude incidence of COVID‐19‐related hospital admission among different rheumatologic diseases, with SS being one of the conditions with a higher risk. This finding may support the implication of viral infection in the pathophysiology of SS. Therefore, I propose that autoimmunity against the salivary glands should be carefully monitored, as SARS‐CoV‐2 infects human salivary glands as abundantly so that the saliva could be used as an excellent clinical sample for COVID‐19 diagnosis. 29 The expression of ACE2, the SARS‐CoV‐2 receptor, was even higher in minor salivary glands than in the lungs. 30 After the infection, saliva may cultivate the live virus, and the salivary glands would act as a long‐term viral reservoir of SARS‐CoV‐2. 30 Hence, autoimmune sialoadenitis, which is or at least mimics SS, might become apparent even after susceptible individuals recover from COVID‐19 (Figure 1). The well‐known complaint of altered taste and smell functions in patients with COVID‐19 31 may reflect in a part a clinical manifestation of salivary dysfunction. FIGURE 1 Severe acute respiratory syndrome coronavirus 2 infection with potential of developing Sjögren’s syndrome Conversely, because of the mimicry between viral spike protein and human cells, whether the messenger RNA (mRNA)‐based anti‐SARS‐CoV‐2 vaccines might stimulate an unwanted immune reaction in individuals with immune dysfunctions is controversial. 32 The speculation may not be limited to mRNA vaccines, as an association between specific vaccination (eg, influenza vaccine) and SS or the production of anti‐Ro/anti‐La antibodies has been reported. 23 Thus, the infection and/or vaccination might trigger or accelerate autoimmune responses against the spike protein of the SARS‐CoV‐2 in genetically susceptible individuals or patients with immune dysfunction. In this concern, not only autoimmune rheumatic diseases but also other autoimmunity entities such as hepatitis 33 and thyroiditis 34 have been reported to emerge after vaccination against SARS‐CoV‐2. Therefore, the potential stimulatory effect of SARS‐CoV‐2 vaccines (regardless of mRNA, virus vectored, or protein subunit) to brake immunological tolerance should be examined for both short‐ and long‐term events. Nevertheless, considering the severe outcome of COVID‐19, vaccinations should be processed under a balance between the risk and the benefit for each individual in this pandemic. 32 CONFLICT OF INTEREST None.

          Related collections

          Most cited references38

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID-19

          Objective Although COVID-19 is primarily a respiratory illness, there is mounting evidence suggesting that the GI tract is involved in this disease. We investigated whether the gut microbiome is linked to disease severity in patients with COVID-19, and whether perturbations in microbiome composition, if any, resolve with clearance of the SARS-CoV-2 virus. Methods In this two-hospital cohort study, we obtained blood, stool and patient records from 100 patients with laboratory-confirmed SARS-CoV-2 infection. Serial stool samples were collected from 27 of the 100 patients up to 30 days after clearance of SARS-CoV-2. Gut microbiome compositions were characterised by shotgun sequencing total DNA extracted from stools. Concentrations of inflammatory cytokines and blood markers were measured from plasma. Results Gut microbiome composition was significantly altered in patients with COVID-19 compared with non-COVID-19 individuals irrespective of whether patients had received medication (p<0.01). Several gut commensals with known immunomodulatory potential such as Faecalibacterium prausnitzii, Eubacterium rectale and bifidobacteria were underrepresented in patients and remained low in samples collected up to 30 days after disease resolution. Moreover, this perturbed composition exhibited stratification with disease severity concordant with elevated concentrations of inflammatory cytokines and blood markers such as C reactive protein, lactate dehydrogenase, aspartate aminotransferase and gamma-glutamyl transferase. Conclusion Associations between gut microbiota composition, levels of cytokines and inflammatory markers in patients with COVID-19 suggest that the gut microbiome is involved in the magnitude of COVID-19 severity possibly via modulating host immune responses. Furthermore, the gut microbiota dysbiosis after disease resolution could contribute to persistent symptoms, highlighting a need to understand how gut microorganisms are involved in inflammation and COVID-19.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            TH17 responses in cytokine storm of COVID-19: An emerging target of JAK2 inhibitor Fedratinib

            COVID-19 emerges as a pandemic disease with high mortality. Development of effective prevention and treatment is an urgent need. We reviewed TH17 responses in patients with SARS-CoV-2 and proposed an FDA approved JAK2 inhibitor Fedratinib for reducing mortality of patients with TH17 type immune profiles.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Marked T cell activation, senescence, exhaustion and skewing towards TH17 in patients with COVID-19 pneumonia

              The immune system of patients infected by SARS-CoV-2 is severely impaired. Detailed investigation of T cells and cytokine production in patients affected by COVID-19 pneumonia are urgently required. Here we show that, compared with healthy controls, COVID-19 patients’ T cell compartment displays several alterations involving naïve, central memory, effector memory and terminally differentiated cells, as well as regulatory T cells and PD1+CD57+ exhausted T cells. Significant alterations exist also in several lineage-specifying transcription factors and chemokine receptors. Terminally differentiated T cells from patients proliferate less than those from healthy controls, whereas their mitochondria functionality is similar in CD4+ T cells from both groups. Patients display significant increases of proinflammatory or anti-inflammatory cytokines, including T helper type-1 and type-2 cytokines, chemokines and galectins; their lymphocytes produce more tumor necrosis factor (TNF), interferon-γ, interleukin (IL)-2 and IL-17, with the last observation implying that blocking IL-17 could provide a novel therapeutic strategy for COVID-19.
                Bookmark

                Author and article information

                Contributors
                k_msk@mac.com
                Journal
                Int J Rheum Dis
                Int J Rheum Dis
                10.1111/(ISSN)1756-185X
                APL
                International Journal of Rheumatic Diseases
                John Wiley and Sons Inc. (Hoboken )
                1756-1841
                1756-185X
                22 June 2021
                July 2021
                22 June 2021
                : 24
                : 7 ( doiID: 10.1111/apl.v24.7 )
                : 963-965
                Affiliations
                [ 1 ] Department of Internal Medicine Akasaka Sanno Medical Center Tokyo Japan
                Author notes
                [*] [* ] Correspondence

                Kayo Masuko, Department of Internal Medicine, Akasaka Sanno Medical Center, W building 4‐1‐26, Akasaka, Minato‐ku, Tokyo 107‐8402, Japan.

                Email: k_msk@ 123456mac.com

                Author information
                https://orcid.org/0000-0001-6058-6363
                Article
                APL14154
                10.1111/1756-185X.14154
                8441922
                34156141
                9491cc9c-e8d5-4c68-9b36-6de683112462
                © 2021 Asia Pacific League of Associations for Rheumatology and John Wiley & Sons Australia, Ltd.

                This article is being made freely available through PubMed Central as part of the COVID-19 public health emergency response. It can be used for unrestricted research re-use and analysis in any form or by any means with acknowledgement of the original source, for the duration of the public health emergency.

                History
                : 01 May 2021
                : 01 March 2021
                : 20 May 2021
                Page count
                Figures: 1, Tables: 0, Pages: 3, Words: 2089
                Categories
                Novel Hypothesis
                Novel Hypothesis
                Custom metadata
                2.0
                July 2021
                Converter:WILEY_ML3GV2_TO_JATSPMC version:6.0.7 mode:remove_FC converted:15.09.2021

                Rheumatology
                autoimmune diseases,autoimmunity,coronavirus disease 2019,sjögren’s syndrome,systemic lupus erythematosus

                Comments

                Comment on this article