1
|
Hansen N, Buschatzky V, Bastin AK, Rentzsch K, Teegen B, Luedecke D, Skripuletz T, Maier HB, Bleich S, Gallinat J, Esselmann H, Dunay IR, Zerr I, Fitzner D, Wilftang J, Neyazi A, Schott BH, Malchow B. Neural autoantibodies in psychiatric disorders are associated with antibodies against viral pathogens: a retrospective study of 619 patients. J Neural Transm (Vienna) 2025:10.1007/s00702-025-02943-x. [PMID: 40382511 DOI: 10.1007/s00702-025-02943-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/03/2025] [Indexed: 05/20/2025]
Abstract
A history of viral infection has been associated with a higher risk for psychiatric disorders. One potential underlying mechanism is that antiviral immunological responses could trigger cross-reactivity between viral and neural antigens, which would raise the co-occurrence of antiviral antibodies and anti-neural autoantibodies. We studied 619 patients' psychiatric diagnoses from the Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Germany. Anti-neural autoantibodies and antiviral antibody specific indices were measured in serum and/or cerebrospinal fluid (CSF) from all patients. Among these 619 patients, 115 tested positive for serum and/or CSF neural autoantibodies (18.6%), with the most often identified autoantibodies being anti-GAD65 in serum (2.2%) and CSF (1.6%), and anti-NMDA in serum (0.6%) and CSF (1.3%). The three main diagnostic groups presenting neural autoantibodies were patients with organic psychiatric disorders including dementia (81 of 377; 21.7%), those with psychotic disorders (9 of 66; 13.6%), and patients with affective disorders (19 of 138; 13.9%). Logistic regression analysis revealed a significant association between the varicella zoster virus (VZV) antibody-specific index and autoantibody positivity in patients with all diagnoses (F00-F79) (p < 0.005). Furthermore, the rubella antibody-specific index proved to be significantly associated with neural autoantibody positivity (p < 0.001) across all patients (F00-F79), and in those with affective disorders (p < 0.01). Our results show that VZV and rubella antiviral antibodies are associated with a higher propensity to develop anti-neural autoantibodies, suggesting that the known association between viral infection and later developing psychiatric disorders may be partly attributable to the development of anti-neural autoimmunity.
Collapse
Affiliation(s)
- Niels Hansen
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany.
| | - Vincent Buschatzky
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany
| | - Anne Katharina Bastin
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany
| | - Kristin Rentzsch
- Clinical Immunological Laboratory Prof. Stöcker, Groß Grönau, Germany
| | - Bianca Teegen
- Clinical Immunological Laboratory Prof. Stöcker, Groß Grönau, Germany
| | - Daniel Luedecke
- Department of Psychiatry and Psychotherapy, University Hospital Hamburg Eppendorf, Hamburg, Germany
| | | | - Hannah Benedictine Maier
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Stefan Bleich
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Jürgen Gallinat
- Department of Psychiatry and Psychotherapy, University Hospital Hamburg Eppendorf, Hamburg, Germany
| | - Hermann Esselmann
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany
| | - Ildiko Rita Dunay
- Institute for Inflammation and Neurodegeneration, Otto-Von-Guericke-University Magdeburg, Magdeburg, Germany
- Center for Behavioral Brain Sciences, Otto-Von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Inga Zerr
- Department of Neurology, University Medical Center, Göttingen, Germany
| | - Dirk Fitzner
- Department of Neurology, University Medical Center, Göttingen, Germany
| | - Jens Wilftang
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Alexandra Neyazi
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
- Department of Psychiatry and Psychotherapy, Otto-Von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Björn Hendrik Schott
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany
- Center for Behavioral Brain Sciences, Otto-Von-Guericke-University Magdeburg, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany
- Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Berend Malchow
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Von-Siebold-Str. 5, 37075, Göttingen, Germany
| |
Collapse
|
2
|
Miles MA, Huttmann TD, Liong S, Liong F, O’Leary JJ, Brooks DA, Selemidis S. Exploring the Contribution of TLR7 to Sex-Based Disparities in Respiratory Syncytial Virus (RSV)-Induced Inflammation and Immunity. Viruses 2025; 17:428. [PMID: 40143355 PMCID: PMC11946665 DOI: 10.3390/v17030428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
TLR7 plays a key role in recognizing viral RNA to initiate an immune response. Sex-based differences in the severity of RSV respiratory infections have been noted, and this may be related to higher expression of X-linked toll-like receptor 7 (TLR7) in female immune cells. Indeed, TLR7 has been shown to influence sex differences in responses to other respiratory viruses; however, its role in RSV infection remains underexplored. We infected adult C57Bl/6 or TLR7 knockout mice with RSV and compared the specific lung immune responses between different sexes. Gene expression analysis revealed that infected female mice had elevated levels of type I and II interferons, proinflammatory cytokines, chemokines, and viral transcripts in their lungs compared to males. Additionally, females exhibited increased numbers of macrophages and higher antibody responses in the airways. Deletion of TLR7 diminished the sex differences in certain cytokine and antibody responses. Furthermore, ex vivo infection of male alveolar macrophages with RSV resulted in greater production of proinflammatory cytokines and viral transcripts than in female macrophages, suggesting inherent sex differences in macrophage responses. These findings provide new insights into the mechanisms underlying sex differences in RSV pathophysiology and suggest that TLR7 contributes to an enhanced inflammatory response in females.
Collapse
Affiliation(s)
- Mark A. Miles
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia; (M.A.M.); (T.D.H.); (S.L.); (F.L.)
| | - Thomas D. Huttmann
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia; (M.A.M.); (T.D.H.); (S.L.); (F.L.)
| | - Stella Liong
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia; (M.A.M.); (T.D.H.); (S.L.); (F.L.)
| | - Felicia Liong
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia; (M.A.M.); (T.D.H.); (S.L.); (F.L.)
| | - John J. O’Leary
- Discipline of Histopathology, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, D08 XW7X Dublin, Ireland;
- Sir Patrick Dun’s Laboratory, Central Pathology Laboratory, St James’s Hospital, D08 XW7X Dublin, Ireland
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia;
| | - Doug A. Brooks
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia;
| | - Stavros Selemidis
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia; (M.A.M.); (T.D.H.); (S.L.); (F.L.)
| |
Collapse
|
3
|
Kelley K, Dogru D, Huang Q, Yang Y, Palm NW, Altindis E, Ludvigsson J. Children who develop celiac disease are predicted to exhibit distinct metabolic pathways among their gut microbiota years before diagnosis. Microbiol Spectr 2025; 13:e0146824. [PMID: 39902908 PMCID: PMC11878042 DOI: 10.1128/spectrum.01468-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 01/07/2025] [Indexed: 02/06/2025] Open
Abstract
Celiac disease (CD) is an autoimmune disease caused by a loss of gluten tolerance in genetically predisposed individuals. While 30%-40% of people possess the predisposing alleles, only 1%-2% are diagnosed with CD, suggesting that environmental factors are involved in disease pathogenesis. To determine an association between pediatric CD and the gut microbiome, we analyzed fecal samples from a prospective cohort study (ABIS). These samples were collected from children who later developed CD (CD progressors) and age-matched healthy children (at ages 1, 2.5, and 5) with similar HLA genotypes, breastfeeding durations, and gluten exposure times. We previously reported gut microbiome differences at ages 2.5 and 5 in this cohort; here, we present findings from samples collected at age 1 (n = 5). We identified 14 ASVs differing significantly between CD progressors and controls, including taxa linked to CD pathogenesis. CD progressors had increased Firmicutes and higher alpha diversity in IgA- bacteria. Using PICRUSt, we analyzed metabolic pathways enriched in CD progressors compared to controls at ages 1, 2.5, and 5 (n = 5-16), revealing enriched inflammatory and pathogenic pathways potentially contributing to CD-related immune dysregulation. While results are based on the primary EdgeR analysis, we also applied a non-parametric method of statistical analysis, reporting those results with supplementary figures. In conclusion, our findings suggest distinct metabolic pathways enriched in the gut microbiome of CD progressors years before diagnosis, which could inform targeted therapeutics for CD. As discussed in the limitations section, this small pilot study should be replicated with larger sample sizes for broader generalization. IMPORTANCE We analyzed gut microbiome data from children who later developed celiac disease (CD progressors) compared to healthy children in the first 5 years of life. Using fecal samples corresponding to the three phases of gut microbiome development, we uncovered enriched functional microbial pathways in CD progressors at age 1. Some of these pathways, implicated in bacterial pathogenesis, microbiota modulation, and inflammation, have been correlated with CD. We also identified taxa in CD progressors at age 1 including Lachnospiraceae, Alistipes, and Bifidobacterium dentium that were previously associated with CD. These findings suggest a potential role for these taxa and enriched pathways in pediatric CD onset years before diagnosis, highlighting potential for early interventions. While the findings of this exploratory study should be validated with larger sample sizes, our study suggests microbial metabolic pathways related to CD onset, enhancing our understanding of CD pathogenesis and the role of gut microbiome-mediated early alterations.
Collapse
Affiliation(s)
- Kristina Kelley
- Boston College Biology Department, Chestnut Hill, Massachusetts, USA
| | - Dogus Dogru
- Boston College Biology Department, Chestnut Hill, Massachusetts, USA
| | - Qian Huang
- Boston College Biology Department, Chestnut Hill, Massachusetts, USA
| | - Yi Yang
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Noah W. Palm
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Emrah Altindis
- Boston College Biology Department, Chestnut Hill, Massachusetts, USA
| | - Johnny Ludvigsson
- Crown Princess Victoria’s Children’s Hospital, Region Östergötland, Linköping, Sweden
- Division of Pediatrics, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
4
|
Borghol AH, Bitar ER, Hanna A, Naim G, Rahal EA. The role of Epstein-Barr virus in autoimmune and autoinflammatory diseases. Crit Rev Microbiol 2025; 51:296-316. [PMID: 38634723 DOI: 10.1080/1040841x.2024.2344114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/15/2024] [Accepted: 04/11/2024] [Indexed: 04/19/2024]
Abstract
Epstein-Barr Virus (EBV), a dsDNA herpesvirus, is believed to play a significant role in exacerbating and potentially triggering autoimmune and autoinflammatory maladies. Around 90% of the world is infected with the virus, which establishes latency within lymphocytes. EBV is also known to cause infectious mononucleosis, a self-limited flu-like illness, in adolescents. EBV is often reactivated and it employs several mechanisms of evading the host immune system. It has also been implicated in inducing host immune dysfunction potentially resulting in exacerbation or triggering of inflammatory processes. EBV has therefore been linked to a number of autoimmune diseases, including systemic lupus erythematosus, multiple sclerosis, rheumatoid arthritis, and Sjögren's syndrome. The review examines the molecular mechanisms through which the virus alters host immune system components thus possibly resulting in autoimmune processes. Understanding the mechanisms underpinning EBV-associated autoimmunity is pivotal; however, the precise causal pathways remain elusive. Research on therapeutic agents and vaccines for EBV has been stagnant for a long number of years until recent advances shed light on potential therapeutic targets. The implications of EBV in autoimmunity underscore the importance of developing targeted therapeutic strategies and, potentially, vaccines to mitigate the autoimmune burden associated with this ubiquitous virus.
Collapse
Affiliation(s)
- Abdul Hamid Borghol
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Center for Infectious Diseases Research (CIDR), American University of Beirut, Beirut, Lebanon
| | - Elio R Bitar
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Center for Infectious Diseases Research (CIDR), American University of Beirut, Beirut, Lebanon
| | - Aya Hanna
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Center for Infectious Diseases Research (CIDR), American University of Beirut, Beirut, Lebanon
| | - Georges Naim
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Center for Infectious Diseases Research (CIDR), American University of Beirut, Beirut, Lebanon
| | - Elias A Rahal
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Center for Infectious Diseases Research (CIDR), American University of Beirut, Beirut, Lebanon
| |
Collapse
|
5
|
Chen X, Ghanizada M, Mallajosyula V, Sola E, Capasso R, Kathuria KR, Davis MM. Differential roles of human CD4 + and CD8 + regulatory T cells in controlling self-reactive immune responses. Nat Immunol 2025; 26:230-239. [PMID: 39806065 PMCID: PMC11785521 DOI: 10.1038/s41590-024-02062-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025]
Abstract
Here we analyzed the relative contributions of CD4+ regulatory T cells expressing Forkhead box protein P3 (FOXP3) and CD8+ regulatory T cells expressing killer cell immunoglobulin-like receptors to the control of autoreactive T and B lymphocytes in human tonsil-derived immune organoids. FOXP3 and GZMB respectively encode proteins FOXP3 and granzyme B, which are critical to the suppressive functions of CD4+ and CD8+ regulatory T cells. Using CRISPR-Cas9 gene editing, we were able to achieve a reduction of ~90-95% in the expression of these genes. FOXP3 knockout in tonsil T cells led to production of antibodies against a variety of autoantigens and increased the affinity of influenza-specific antibodies. By contrast, GZMB knockout resulted in an increase in follicular helper T cells, consistent with the ablation of CD8+ regulatory T cells observed in mouse models, and a marked expansion of autoreactive CD8+ and CD4+ T cells. These findings highlight the distinct yet complementary roles of CD8+ and CD4+ regulatory T cells in regulating cellular and humoral responses to prevent autoimmunity.
Collapse
Affiliation(s)
- Xin Chen
- Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, CA, USA
| | - Mustafa Ghanizada
- Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, CA, USA
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Vamsee Mallajosyula
- Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, CA, USA
| | - Elsa Sola
- Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, CA, USA
| | - Robson Capasso
- Division of Sleep Surgery, Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Karan Raj Kathuria
- Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, CA, USA
| | - Mark M Davis
- Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, CA, USA.
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA.
- The Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
6
|
Ezzedine K, Tannous R, Pearson TF, Harris JE. Recent clinical and mechanistic insights into vitiligo offer new treatment options for cell-specific autoimmunity. J Clin Invest 2025; 135:e185785. [PMID: 39817457 PMCID: PMC11735104 DOI: 10.1172/jci185785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025] Open
Abstract
Vitiligo is an autoimmune disease that has been recognized, stigmatized, and treated for millennia. Recent translational research has revealed key mechanisms of disease, including cellular stress, innate immune activation, T cell-mediated elimination of melanocytes from the skin resulting in clinically apparent white spots, as well as stem cell regeneration that reverses established lesions. Many of these pathways have been targeted therapeutically, leading to the first FDA-approved medication to reverse the disease, with many more in clinical trials. Despite these impressive advances, many questions remain, which will be answered through integration of additional basic, translational, and clinical research studies. This vitiligo revolution has led to great excitement for individuals with vitiligo, those who know them, and the dermatologists who care for their patients. But just as importantly, these advances have great potential to shed light on autoimmune diseases that are more difficult to study, possibly leading to treatment advances that could not be achieved otherwise.
Collapse
Affiliation(s)
- Khaled Ezzedine
- Department of Dermatology, Hôpital Henri Mondor, Université Paris-Est Créteil Val de Marne-Université Paris, Paris, France
- EpidermE, Université Paris-Est Créteil (UPEC), Créteil, France
| | - Rim Tannous
- Department of Dermatology, Hôpital Henri Mondor, Université Paris-Est Créteil Val de Marne-Université Paris, Paris, France
| | - Todd F. Pearson
- Department of Dermatology, UMass Chan Medical School, Worcester, Massachusetts, USA
| | - John E. Harris
- Department of Dermatology, UMass Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
7
|
Zhou F, Xu Y, Liu X, Xu Y, Wang Y, Jiang D, Du P. Zika and Dengue Virus Autoimmunity: An Overview of Related Disorders and Their Potential Mechanisms. Rev Med Virol 2025; 35:e70014. [PMID: 39779915 DOI: 10.1002/rmv.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025]
Abstract
Zika virus (ZIKV) and dengue virus (DENV) are two major mosquito-borne flaviviruses that pose a significant threat to the global public health system, particularly in tropical regions. The clinical outcomes related to these viral pathogens can vary from self-limiting asymptomatic infections to various forms of life-threatening pathological conditions such as haemorrhagic disorders. In addition to the direct effects of the viral pathogens, immune processes play also a significant function in the development of diseases mediated by ZIKV and DENV. Studing these processes is important for developing safer vaccines and targeted therapeutic strategies. These viruses have been reported to trigger various autoimmune disorders affecting different parts of human organ systems. It also has been shown that preexisting immunity to ZIKV or DENV can impact the outcome of subsequent infections caused by another virus. ZIKV and DENV infection can promote the development of autoimmune disorders by different mechanisms, such as molecular mimicry and autoantibody formation. The present review provides an overview of various autoimmune disorders associated with ZIKV and DENV infection and their potential underlying mechanisms.
Collapse
Affiliation(s)
- Feifan Zhou
- Department of Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yuanze Xu
- The Second Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xing Liu
- Department of Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yan Xu
- Department of Stomach Enterochirurgia, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yan Wang
- Department of Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Donghui Jiang
- Department of Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Pengfei Du
- Department of Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
8
|
Hu P, Li H, Ji Z, Jing W, Li Z, Yu S, Shan X, Cui Y, Wang B, Dong H, Zhou Y, Wang Z, Xiong H, Zhang X, Li HC, Wang J, Tang J, Wang T, Xie K, Liu Y, Zhu H, Yu Q. Fructose-1,6-diphosphate inhibits viral replication by promoting the lysosomal degradation of HMGB1 and blocking the binding of HMGB1 to the viral genome. PLoS Pathog 2024; 20:e1012782. [PMID: 39693295 DOI: 10.1371/journal.ppat.1012782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024] Open
Abstract
Fructose-1,6-diphosphate (FBP), a key glycolytic metabolite, is recognized for its cytoprotective effects during stress. However, the role of FBP in viral infections is unknown. Here, we demonstrate that virus-infected cells exhibit elevated FBP levels. Exogenous FBP inhibits both RNA and DNA virus infections in vitro and in vivo. Modulating intracellular FBP levels by regulating the expression of the metabolic enzymes FBP1 and PFK1 significantly impacts viral infections. Mechanistically, the inhibitory effects of FBP are not a result of altered viral adhesion or entry and are largely independent of type I interferon-mediated immune responses; rather, they occur through modulation of HMGB1. During viral infections, FBP predominantly reduces the protein levels of HMGB1 by facilitating its lysosomal degradation. Furthermore, FBP interacts with HMGB1 and disrupts the binding of HMGB1 to viral genomes, thereby further inhibiting viral replication. Our findings underscore the potential of FBP as a therapeutic target for controlling viral infections.
Collapse
Affiliation(s)
- Penghui Hu
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Huiyi Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Department of Pathogen Biology, School of Basic Medicine and Life Science, Department of Clinical Laboratory of the Second Affiliated Hospital of Hainan Medical University, The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University, Hainan, China
| | - Zemin Ji
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Weijia Jing
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Zihan Li
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Sujun Yu
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xiao Shan
- Department of Health Management Centre & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yan Cui
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Baochen Wang
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hongyuan Dong
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yanzhao Zhou
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
- University of Electronic Science and Technology of China, Chengdu, China
| | - Zhe Wang
- Department of Health Management Centre & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Hui Xiong
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xiaomei Zhang
- Department of Health Management Centre & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Hui-Chieh Li
- Department of Health Management Centre & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jinrong Wang
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jiuzhou Tang
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ting Wang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, Tianjin Medical University, Tianjin, China
| | - Keliang Xie
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuping Liu
- Department of Health Management Centre & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Haizhen Zhu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Department of Pathogen Biology, School of Basic Medicine and Life Science, Department of Clinical Laboratory of the Second Affiliated Hospital of Hainan Medical University, The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University, Hainan, China
| | - Qiujing Yu
- Department of Health Management Centre & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
9
|
Li Y, Xiao J, Li C, Yang M. Memory inflation: Beyond the acute phase of viral infection. Cell Prolif 2024; 57:e13705. [PMID: 38992867 PMCID: PMC11628752 DOI: 10.1111/cpr.13705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 07/13/2024] Open
Abstract
Memory inflation is confirmed as the most commonly dysregulation of host immunity with antigen-independent manner in mammals after viral infection. By generating large numbers of effector/memory and terminal differentiated effector memory CD8+ T cells with diminished naïve subsets, memory inflation is believed to play critical roles in connecting the viral infection and the onset of multiple diseases. Here, we reviewed the current understanding of memory inflated CD8+ T cells in their distinct phenotypic features that different from exhausted subsets; the intrinsic and extrinsic roles in regulating the formation of memory inflation; and the key proteins in maintaining the expansion and proliferation of inflationary populations. More importantly, based on the evidences from both clinic and animal models, we summarized the potential mechanisms of memory inflation to trigger autoimmune neuropathies, such as Guillain-Barré syndrome and multiple sclerosis; the correlations of memory inflation between tumorigenesis and resistance of tumour immunotherapies; as well as the effects of memory inflation to facilitate vascular disease progression. To sum up, better understanding of memory inflation could provide us an opportunity to beyond the acute phase of viral infection, and shed a light on the long-term influences of CD8+ T cell heterogeneity in dampen host immune homeostasis.
Collapse
Affiliation(s)
- Yanfei Li
- School of Basic Medical SciencesChengdu University of Traditional Chinese MedicineChengduChina
| | - Jie Xiao
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Chen Li
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Mu Yang
- School of Basic Medical SciencesChengdu University of Traditional Chinese MedicineChengduChina
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| |
Collapse
|
10
|
Duan L, Li S, Chen D, Shi Y, Zhou X, Feng Y. Causality between autoimmune diseases and schizophrenia: a bidirectional Mendelian randomization study. BMC Psychiatry 2024; 24:817. [PMID: 39550571 PMCID: PMC11568594 DOI: 10.1186/s12888-024-06287-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 11/11/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND Observational studies have shown a link between autoimmune diseases and schizophrenia, with conflicting conclusions. Due to the existence of confounding factors, the causal link between autoimmune diseases and schizophrenia is still unknown. METHOD We conducted a comprehensive Mendelian randomization (MR) analysis of schizophrenia and ten common autoimmune diseases in individuals of European descent using genome-wide association studies (GWASs). To evaluate the relationships between autoimmune diseases and schizophrenia, inverse variance weighted, MR-RAPS, Bayesian weighted MR, constrained maximum likelihood, debiased IVW, MR-Egger, and weighted median were utilized. Several sensitivity analyses were performed to ensure the reliability of the study's results. RESULTS Our findings reveal that genetically predicted ankylosing spondylitis is related to an increased risk of schizophrenia, whereas celiac disease, type 1 diabetes, and systemic lupus erythematosus are associated with a lower risk of schizophrenia. In the reverse MR analysis, our study indicated that genetically predicted schizophrenia is linked to higher risks of ankylosing spondylitis, Crohn's disease, ulcerative colitis, inflammatory bowel disease, and psoriasis. Neither multiple sclerosis nor rheumatoid arthritis have been linked to schizophrenia, and vice versa. CONCLUSION Despite contradicting some other observational reports, this study showed support for a causal link between autoimmune diseases and schizophrenia. To gain a better understanding of the mechanisms underlying the development of immune-mediated schizophrenia, additional research is required to identify potential mechanisms identified in observational studies.
Collapse
Affiliation(s)
- Lincheng Duan
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shiyin Li
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dongnan Chen
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yue Shi
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xianhua Zhou
- Meishan Hospital of Traditional Chinese Medicine, Affiliated Meishan Hospital of Chengdu University of Traditional Chinese Medicine, Meishan, China.
| | - Yue Feng
- Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
11
|
Su Y, Wang J, Ren Y, Xu S, Si Y, Tang M, Li Y, Wang M. Epidemiological Characteristics of Neuro-Specific Antibodies Following Viral Infections. J Med Virol 2024; 96:e70050. [PMID: 39540343 DOI: 10.1002/jmv.70050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/13/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
This study aims to explore the epidemiological characteristics of neuro-specific antibodies (ns-Ab) induced by different viral infections within the central nervous system (CNS). Additionally, it seeks to compare the autoimmune effects following several typical viral infections in CNS. We conduct a retrospective study to compare and analyze the prevalence trends of ns-Ab in patients with different viral infections. Additionally, evaluate the intensity of CNS inflammatory responses postviral infection by correlating clinical characteristics and laboratory findings, and briefly demonstrate the immune effects in CNS following various viral infections. This study retrospectively collected data from 1037 patients hospitalized with suspected CNS infections. A total of 654 patients (63.1%) were included in the final analysis. A higher proportion of patients with pathogens present in their cerebrospinal fluid (CSF) (114 out of 332, 34.3%) tested positive for ns-Ab compared to those without pathogens (70 out of 322, 21.7%) (p = 0.0004). Specifically, the screening rate for ns-Ab in patients with CNS viral infections (83 out of 165, 50.3%) and the prevalence of ns-Ab (27 out of 83, 32.5%) were significantly higher than in those with other pathogen infections (p < 0.0001 and p = 0.016, respectively). Among these, human herpesvirus 7 (HHV7) patients had the highest detection rate of ns-Ab during the disease course (11 out of 26, 42.3%), but exhibited infection characteristics distinctly different from those of herpes simplex virus 1 (HSV1). Viral infections significantly promote the development of autoimmune responses in CNS. The production of ns-Ab and the subsequent autoimmune response vary across different viral infections. There is a strong statistical correlation between HHV7 and the presence of ns-Ab, suggesting that HHV7 may serve as an early indicator of secondary autoimmune response following CNS infections.
Collapse
Affiliation(s)
- Yang Su
- Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Jierui Wang
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yan Ren
- Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Songtao Xu
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yanjun Si
- Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Meng Tang
- Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yi Li
- Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Minjin Wang
- Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
12
|
Nielsen TD, Laursen TM, Bech BH, Rasmussen MM. Traumatic spinal cord injury and its correlation to risk of autoimmune/-inflammatory disease. Spinal Cord 2024; 62:642-650. [PMID: 39261594 PMCID: PMC11549038 DOI: 10.1038/s41393-024-01026-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/14/2024] [Accepted: 08/29/2024] [Indexed: 09/13/2024]
Abstract
STUDY DESIGN Nationwide epidemiological open cohort study. OBJECTIVES To evaluate whether individuals with traumatic spinal cord injury (TSCI) are more prone to develop autoimmune diseases compared to a general non-TSCI population. SETTING Danish public national registries. METHODS An open nationwide cohort, including individuals born in Denmark from or alive during 1945-2018 was collected and the study period was 1980-2018. Poissons Log-linear regression estimated the incidence rate ratio (IRR) for developing eight groups of autoimmune diseases. A dose-response relationship based on the cervical/thoracic level of injury was assessed by stratification. RESULTS The cohort included 3,272 individuals with TSCI and 4.8 million background individuals, accounting for 50,865 and 140 million person-years respectively. The TSCI population had an overall IRR of 1.81 (95% CI, 1.59 to 2.05) of getting any autoimmune disease. Subgroup analysis found positive associations for; a) Other neurologic IRR 5.19 (95% CI, 2.79 to 9.65), b) multiple sclerosis IRR 3.70 (95% CI, 2.54 to 5.40), c) Dermatologic IRR 2.57 (95% CI, 1.86 to 3.55), d) Type 1 diabetes mellitus IRR 2.01 (95% CI, 1.54 to 2.61), e) Systemic 1.92 (95% CI, 1.44 to 2.55), and f) Gastroenterologic IRR 1.42 (95% CI, 1.05 to 1.92). Cervical levels of TSCI showed an IRR of 1.70 (95% CI, 1.43 to 2.02), while thoracic levels had an IRR 1.98 (95% CI, 1.63 to 2.39). CONCLUSIONS TSCI may be an individual risk factor of developing an autoimmune disease. There does not appear to exist a dose-response relationship from the level of injury. SPONSORSHIP None.
Collapse
Affiliation(s)
- Tim Damgaard Nielsen
- Cense Spine, Department of Neurosurgery, Aarhus University Hospital, Aarhus N, Denmark.
- Department of Clinical medicine, Aarhus University, Aarhus N, Denmark.
| | - Thomas Munk Laursen
- Department of Economics and Business economics, Aarhus University, Aarhus V, Denmark
| | - Bodil Hammer Bech
- Department of Public Health, Aarhus University, aarhus, Denmark
- Department of Epidemiology, Aarhus University, Aarhus, Denmark
| | - Mikkel Mylius Rasmussen
- Cense Spine, Department of Neurosurgery, Aarhus University Hospital, Aarhus N, Denmark
- Department of Clinical medicine, Aarhus University, Aarhus N, Denmark
| |
Collapse
|
13
|
Grandinetti R, Mussi N, Pilloni S, Ramundo G, Miniaci A, Turco E, Piccolo B, Capra ME, Forestiero R, Laudisio S, Boscarino G, Pedretti L, Menoni M, Pellino G, Tagliani S, Bergomi A, Antodaro F, Cantù MC, Bersini MT, Mari S, Mazzini F, Biasucci G, Suppiej A, Esposito S. Pediatric acute-onset neuropsychiatric syndrome and pediatric autoimmune neuropsychiatric disorder associated with streptococcal infections: a delphi study and consensus document about definition, diagnostic criteria, treatment and follow-up. Front Immunol 2024; 15:1420663. [PMID: 39512340 PMCID: PMC11540630 DOI: 10.3389/fimmu.2024.1420663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 10/07/2024] [Indexed: 11/15/2024] Open
Abstract
Pediatric Autoimmune Neuropsychiatric Disorder Associated with Streptococcal Infections (PANDAS) and Pediatric Acute-onset Neuropsychiatric Syndrome (PANS) are broad diagnoses that encompass a range of sudden-onset neuropsychiatric symptoms in children, which can include obsessive-compulsive disorder (OCD), tics, anxiety, emotional instability, and cognitive difficulties. Unlike PANDAS, PANS is not strictly linked to group A streptococcal infections but can be triggered by various infectious or environmental factors. Lights and shadows remain upon the management of children with PANS and PANDAS and there is no clear consensus regarding definition, diagnostic criteria, treatment, and follow-up. The aim of the present study was to evaluate the level of agreement on PANS and PANDAS definition, diagnostic criteria, treatment and follow-up and to assess on the basis of recent studies whether there is a need to modify the current recommendations used by primary care pediatricians and hospital pediatricians in clinical practice in order to improve outcomes. Using the Delphi method, this consensus provides shared indications on PANS and PANDAS management in pediatric age, based on the most updated literature. This work represents, in our opinion, the most complete and up-to-date information on the diagnosis of PANS and PANDAS, as well as consensus statements about several aspects of clinical care. Undoubtedly, more randomized and controlled trials are needed in the pediatric population to better define the best management, also in terms of adequate follow-up examinations and period of observation.
Collapse
Affiliation(s)
- Roberto Grandinetti
- Pediatric Clinic, University Hospital, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Nicole Mussi
- Pediatric Clinic, University Hospital, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Simone Pilloni
- Pediatric Clinic, University Hospital, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Greta Ramundo
- Pediatric Clinic, University Hospital, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Angela Miniaci
- Pediatric Clinic, IRCCS Azienda Ospedaliera Universitaria di Bologna, Bologna, Italy
| | - Emanuela Turco
- Pediatric Clinic, University Hospital, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Benedetta Piccolo
- Pediatric Clinic, University Hospital, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Maria Elena Capra
- Pediatrics and Neonatology Unit, Department of Medicine and Surgery, University of Parma, Guglielmo da Saliceto Hospital, Piacenza, Italy
| | - Roberta Forestiero
- Pediatric Clinic, University Hospital, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Serena Laudisio
- Pediatric Clinic, University Hospital, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Giovanni Boscarino
- Pediatric Clinic, University Hospital, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Laura Pedretti
- Pediatric Clinic, University Hospital, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Martina Menoni
- Pediatric Clinic, University Hospital, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | | | - Andrea Bergomi
- Primary Care Pediatricians, Azienda Unit Sanitaria Locale (AUSL) Modena, Modena, Italy
| | - Francesco Antodaro
- Primary Care Pediatricians, Azienda Unit Sanitaria Locale (AUSL) Modena, Modena, Italy
| | - Maria Cristina Cantù
- Primary Care Pediatricians, Azienda Unit Sanitaria Locale (AUSL) Parma, Parma, Italy
| | - Maria Teresa Bersini
- Primary Care Pediatricians, Azienda Unit Sanitaria Locale (AUSL) Parma, Parma, Italy
| | - Sandra Mari
- Primary Care Pediatricians, Azienda Unit Sanitaria Locale (AUSL) Parma, Parma, Italy
| | - Franco Mazzini
- Primary Care Pediatricians, Azienda Unit Sanitaria Locale (AUSL) Romagna, Forlì-Cesena, Italy
| | - Giacomo Biasucci
- Pediatrics and Neonatology Unit, Department of Medicine and Surgery, University of Parma, Guglielmo da Saliceto Hospital, Piacenza, Italy
| | | | - Susanna Esposito
- Pediatric Clinic, University Hospital, Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
14
|
Ayoub I, Freeman SA, Saoudi A, Liblau R. Infection, vaccination and narcolepsy type 1: Evidence and potential molecular mechanisms. J Neuroimmunol 2024; 393:578383. [PMID: 39032452 DOI: 10.1016/j.jneuroim.2024.578383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/13/2024] [Accepted: 05/31/2024] [Indexed: 07/23/2024]
Abstract
NT1 is a rare, chronic and disabling neurological disease causing excessive daytime sleepiness and cataplexy. NT1 is characterized pathologically by an almost complete loss of neurons producing the hypocretin (HCRT)/orexin neuropeptides in the lateral hypothalamus. While the exact etiology of NT1 is still unknown, numerous studies have provided compelling evidence supporting its autoimmune origin. The prevailing hypothetical view on the pathogenesis of NT1 involves an immune-mediated loss of HCRT neurons that can be triggered by Pandemrix® vaccination and/or by infection in genetically susceptible patients, specifically carriers of the HLA-DQB1*06:02 MHC class II allele. The molecular mechanisms by which infection/vaccination can induce autoimmunity in the case of NT1 remain to be elucidated. In this review, evidence regarding the involvement of vaccination and infection and the potential mechanisms by which it could be linked to the pathogenesis of NT1 will be discussed in light of the existing findings in other autoimmune diseases.
Collapse
Affiliation(s)
- Ikram Ayoub
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, Toulouse, France.
| | - Sean A Freeman
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, Toulouse, France; Department of Neurology, Toulouse University Hospitals, Toulouse, France
| | - Abdelhadi Saoudi
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, Toulouse, France
| | - Roland Liblau
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, Toulouse, France; Department of Immunology, Toulouse University Hospitals, Toulouse, France
| |
Collapse
|
15
|
Sutanto H, Safira A, Fetarayani D. From tumor to tolerance: A comprehensive review of immune checkpoint inhibitors and immune-related adverse events. Asia Pac Allergy 2024; 14:124-138. [PMID: 39220570 PMCID: PMC11365684 DOI: 10.5415/apallergy.0000000000000146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 05/02/2024] [Indexed: 09/04/2024] Open
Abstract
The advent of immune checkpoint inhibitors (ICIs) has revolutionized the treatment landscape for various malignancies by harnessing the body's immune system to target cancer cells. However, their widespread use has unveiled a spectrum of immune-related adverse events, highlighting a critical balance between antitumor immunity and autoimmunity. This review article delves into the molecular immunology of ICIs, mapping the journey from their therapeutic action to the unintended induction of immune-related adverse events. We provide a comprehensive overview of all available ICIs, including cytotoxic T-lymphocyte-associated protein 4, programmed cell death protein 1, programmed death-ligand 1 inhibitors, and emerging targets, discussing their mechanisms of action, clinical applications, and the molecular underpinnings of associated immune-related adverse events. Special attention is given to the activation of autoreactive T cells, B cells, cytokine release, and the inflammatory cascade, which together contribute to the development of immune-related adverse events. Through a molecular lens, we explore the clinical manifestations of immune-related adverse events across organ systems, offering insights into diagnosis, management, and strategies to mitigate these adverse effects. The review underscores the importance of understanding the delicate interplay between enhancing antitumor responses and minimizing immune-related adverse events, aiming to guide future research and the development of next-generation ICIs with improved drug safety profiles.
Collapse
Affiliation(s)
- Henry Sutanto
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Ardea Safira
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Deasy Fetarayani
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
16
|
Rodriguez‐Nava G, El Kamari V, Chang H, Egoryan G, Bonilla HF. New-onset sarcoidosis in a patient with long COVID. Clin Case Rep 2024; 12:e9186. [PMID: 39130813 PMCID: PMC11316136 DOI: 10.1002/ccr3.9186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 06/04/2024] [Accepted: 07/09/2024] [Indexed: 08/13/2024] Open
Abstract
Long COVID, often following SARS-CoV-2 infection, may stem from sustained inflammation, overlapping with autoimmune diseases like sarcoidosis. Though specific treatments lack, this link could shape future diagnostic and therapeutic methods.
Collapse
Affiliation(s)
- Guillermo Rodriguez‐Nava
- Division of Infectious Diseases and Geographic Medicine, Department of MedicineStanford University School of MedicineStanfordCaliforniaUSA
| | - Vanessa El Kamari
- Division of Infectious Diseases and Geographic Medicine, Department of MedicineStanford University School of MedicineStanfordCaliforniaUSA
| | - Harvey Chang
- Department of PathologyMemorial Medical CenterModestoCaliforniaUSA
| | - Goar Egoryan
- Division of Oncology, Department of MedicineStanford University School of MedicineStanfordCaliforniaUSA
| | - Hector F. Bonilla
- Division of Infectious Diseases and Geographic Medicine, Department of MedicineStanford University School of MedicineStanfordCaliforniaUSA
- Stanford Post‐Acute COVID‐19 Syndrome ClinicStanford Health CareStanfordCaliforniaUSA
- Stanford Myalgic Encephalomyelitis/Chronic Fatigue Syndrome ClinicStanford Health CareStanfordCaliforniaUSA
| |
Collapse
|
17
|
Zhang H, Wang J, Zhang K, Shi J, Gao Y, Zheng J, He J, Zhang J, Song Y, Zhang R, Shi X, Jin L, Li H. Association between heavy metals exposure and persistent infections: the mediating role of immune function. Front Public Health 2024; 12:1367644. [PMID: 39104887 PMCID: PMC11298456 DOI: 10.3389/fpubh.2024.1367644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 07/11/2024] [Indexed: 08/07/2024] Open
Abstract
Introduction Persistent infections caused by certain viruses and parasites have been associated with multiple diseases and substantial mortality. Heavy metals are ubiquitous environmental pollutants with immunosuppressive properties. This study aimed to determine whether heavy metals exposure suppress the immune system, thereby increasing the susceptibility to persistent infections. Methods Using data from NHANES 1999-2016, we explored the associations between heavy metals exposure and persistent infections: Cytomegalovirus (CMV), Epstein-Barr Virus (EBV), Hepatitis C Virus (HCV), Herpes Simplex Virus Type-1 (HSV-1), Toxoplasma gondii (T. gondii), and Toxocara canis and Toxocara cati (Toxocara spp.) by performing logistic regression, weighted quantile sum (WQS) and Bayesian kernel machine regression (BKMR) models. Mediation analysis was used to determine the mediating role of host immune function in these associations. Results Logistic regression analysis revealed positive associations between multiple heavy metals and the increased risk of persistent infections. In WQS models, the heavy metals mixture was associated with increased risks of several persistent infections: CMV (OR: 1.58; 95% CI: 1.17, 2.14), HCV (OR: 2.94; 95% CI: 1.68, 5.16), HSV-1 (OR: 1.25; 95% CI: 1.11, 1.42), T. gondii (OR: 1.97; 95% CI: 1.41, 2.76), and Toxocara spp. (OR: 1.76; 95% CI: 1.16, 2.66). BKMR models further confirmed the combined effects of heavy metals mixture and also identified the individual effect of arsenic, cadmium, and lead. On mediation analysis, the systemic immune inflammation index, which reflects the host's immune status, mediated 12.14% of the association of mixed heavy metals exposure with HSV-1 infection. Discussion The findings of this study revealed that heavy metals exposure may increase susceptibility to persistent infections, with the host's immune status potentially mediating this relationship. Reducing exposure to heavy metals may have preventive implications for persistent infections, and further prospective studies are needed to confirm these findings.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Lina Jin
- School of Public Health, Jilin University, Changchun, China
| | - Hui Li
- School of Public Health, Jilin University, Changchun, China
| |
Collapse
|
18
|
Duarte LF, Villalobos V, Farías MA, Rangel-Ramírez MA, González-Madrid E, Navarro AJ, Carbone-Schellman J, Domínguez A, Alvarez A, Riedel CA, Bueno SM, Kalergis AM, Cáceres M, González PA. Asymptomatic herpes simplex virus brain infection elicits cellular senescence phenotypes in the central nervous system of mice suffering multiple sclerosis-like disease. Commun Biol 2024; 7:811. [PMID: 38965360 PMCID: PMC11224417 DOI: 10.1038/s42003-024-06486-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/21/2024] [Indexed: 07/06/2024] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a demyelinating disease affecting the central nervous system (CNS) in animals that parallels several clinical and molecular traits of multiple sclerosis in humans. Herpes simplex virus type 1 (HSV-1) infection mainly causes cold sores and eye diseases, yet eventually, it can also reach the CNS, leading to acute encephalitis. Notably, a significant proportion of healthy individuals are likely to have asymptomatic HSV-1 brain infection with chronic brain inflammation due to persistent latent infection in neurons. Because cellular senescence is suggested as a potential factor contributing to the development of various neurodegenerative disorders, including multiple sclerosis, and viral infections may induce a premature senescence state in the CNS, potentially increasing susceptibility to such disorders, here we examine the presence of senescence-related markers in the brains and spinal cords of mice with asymptomatic HSV-1 brain infection, EAE, and both conditions. Across all scenarios, we find a significant increases of senescence biomarkers in the CNS with some differences depending on the analyzed group. Notably, some senescence biomarkers are exclusively observed in mice with the combined conditions. These results indicate that asymptomatic HSV-1 brain infection and EAE associate with a significant expression of senescence biomarkers in the CNS.
Collapse
MESH Headings
- Animals
- Cellular Senescence
- Mice
- Brain/virology
- Brain/pathology
- Brain/metabolism
- Multiple Sclerosis/virology
- Multiple Sclerosis/pathology
- Multiple Sclerosis/metabolism
- Herpesvirus 1, Human/physiology
- Herpesvirus 1, Human/pathogenicity
- Herpes Simplex/virology
- Herpes Simplex/pathology
- Female
- Mice, Inbred C57BL
- Encephalomyelitis, Autoimmune, Experimental/virology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Phenotype
- Central Nervous System/virology
- Central Nervous System/metabolism
- Central Nervous System/pathology
- Spinal Cord/virology
- Spinal Cord/metabolism
- Spinal Cord/pathology
- Biomarkers/metabolism
- Encephalitis, Herpes Simplex/virology
- Encephalitis, Herpes Simplex/pathology
- Encephalitis, Herpes Simplex/metabolism
Collapse
Affiliation(s)
- Luisa F Duarte
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Ciencias Biológicas, Facultad de Ciencias de La Vida, Universidad Andrés Bello, Santiago, Chile
- Centro de Medicina Regenerativa, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Verónica Villalobos
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Mónica A Farías
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ma Andreina Rangel-Ramírez
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Ciencias Biológicas, Facultad de Ciencias de La Vida, Universidad Andrés Bello, Santiago, Chile
| | - Enrique González-Madrid
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Ciencias Biológicas, Facultad de Ciencias de La Vida, Universidad Andrés Bello, Santiago, Chile
| | - Areli J Navarro
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Javier Carbone-Schellman
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Angélica Domínguez
- Departamento de Salud Pública, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandra Alvarez
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A Riedel
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Ciencias Biológicas, Facultad de Ciencias de La Vida, Universidad Andrés Bello, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mónica Cáceres
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
19
|
Tsay GJ, Zouali M. Cellular pathways and molecular events that shape autoantibody production in COVID-19. J Autoimmun 2024; 147:103276. [PMID: 38936147 DOI: 10.1016/j.jaut.2024.103276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/26/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
A hallmark of COVID-19 is the variety of complications that follow SARS-CoV-2 infection in some patients, and that target multiple organs and tissues. Also remarkable are the associations with several auto-inflammatory disorders and the presence of autoantibodies directed to a vast array of antigens. The processes underlying autoantibody production in COVID-19 have not been completed deciphered. Here, we review mechanisms involved in autoantibody production in COVID-19, multisystem inflammatory syndrome in children, and post-acute sequelae of COVID19. We critically discuss how genomic integrity, loss of B cell tolerance to self, superantigen effects of the virus, and extrafollicular B cell activation could underly autoantibody proaction in COVID-19. We also offer models that may account for the pathogenic roles of autoantibodies in the promotion of inflammatory cascades, thromboembolic phenomena, and endothelial and vascular deregulations.
Collapse
Affiliation(s)
- Gregory J Tsay
- Division of Immunology and Rheumatology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan; College of Medicine, China Medical University, Taichung, Taiwan
| | - Moncef Zouali
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.
| |
Collapse
|
20
|
Wang C, Jiang H, Chen S, Zhao Y, Li J, Huang C, Zhou Y, Wang Q, Tian X, Li M, Zeng X, Zhao Y, Wu C, Zhao J. Exploring the impact of acute viral exposure on clinical characteristics and antibody profiles in antiphospholipid syndrome: a study in CAPSTONE. Clin Exp Med 2024; 24:130. [PMID: 38888664 PMCID: PMC11189343 DOI: 10.1007/s10238-024-01400-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024]
Abstract
The relationship between antiphospholipid syndrome (APS) and acute viral infection, such as SARS-CoV-2, is unclear. This study aims to assess symptoms, antiphospholipid antibody (aPL) fluctuations, and complication risks in APS patients infected with SARS-CoV-2. APS patients from Peking Union Medical College Hospital during the COVID-19 outbreak (October-December 2022) were included. Age- and gender-matched APS patients without infection served as controls. Data on demographics, symptoms, treatments, and serum aPL levels were analyzed. Of 234 APS patients, 107 (45.7%) were infected with SARS-CoV-2. Typical symptoms included high fever (81.3%), cough/expectoration (70.1%), and pharyngalgia (52.3%). Age- and gender-based matching selected 97 patients in either infected or uninfected group. After infection, anti-β-2-glycoprotein I-IgG (aβ2GP1-IgG) increased from 4.14 to 4.18 AU/ml, aβ2GP1-IgM decreased from 9.85 to 7.38 AU/ml, and anticardiolipin-IgA (aCL-IgA) significantly increased with a median remaining at 2.50 APLU/ml. Lupus anticoagulants and other aPLs remained stable. Arterial thrombosis incidence increased from 18 (18.6%) to 21 (21.6%), while venous thrombosis incidence did not change. Additionally, 7 (6.5%) patients presented either new-onset or worsening thrombocytopenia, characterized by a significant decline in platelet count (no less than 10 × 109/L) within two weeks of SARS-CoV-2 infection, all of which recovered within 2 weeks. Acute SARS-CoV-2 infection may induce or worsen thrombocytopenia but does not substantially increase thrombotic events in APS. The process of SARS-CoV-2 infection was related to mild titer fluctuation of aβ2GP1-IgG, aβ2GP1-IgM and aCL-IgA in APS patients, necessitating careful monitoring and management.
Collapse
Affiliation(s)
- Chuhan Wang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Hui Jiang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Siyun Chen
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Yuan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Jun Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Can Huang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, 100730, China
- National Clinical Research Center of Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, 100730, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Yangzhong Zhou
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, 100730, China
- National Clinical Research Center of Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, 100730, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Qian Wang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, 100730, China
- National Clinical Research Center of Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, 100730, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Xinping Tian
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, 100730, China
- National Clinical Research Center of Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, 100730, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Mengtao Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, 100730, China
- National Clinical Research Center of Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, 100730, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, 100730, China
- National Clinical Research Center of Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, 100730, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Yan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, 100730, China
- National Clinical Research Center of Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, 100730, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Chuancong Wu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, 100730, China.
- Department of Rheumatology and Immunology, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China.
| | - Jiuliang Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, 100730, China.
- National Clinical Research Center of Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, 100730, China.
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, 100730, China.
| |
Collapse
|
21
|
Rakebrandt N, Yassini N, Kolz A, Schorer M, Lambert K, Goljat E, Estrada Brull A, Rauld C, Balazs Z, Krauthammer M, Carballido JM, Peters A, Joller N. Innate acting memory Th1 cells modulate heterologous diseases. Proc Natl Acad Sci U S A 2024; 121:e2312837121. [PMID: 38838013 PMCID: PMC11181110 DOI: 10.1073/pnas.2312837121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 05/08/2024] [Indexed: 06/07/2024] Open
Abstract
Through immune memory, infections have a lasting effect on the host. While memory cells enable accelerated and enhanced responses upon rechallenge with the same pathogen, their impact on susceptibility to unrelated diseases is unclear. We identify a subset of memory T helper 1 (Th1) cells termed innate acting memory T (TIA) cells that originate from a viral infection and produce IFN-γ with innate kinetics upon heterologous challenge in vivo. Activation of memory TIA cells is induced in response to IL-12 in combination with IL-18 or IL-33 but is TCR independent. Rapid IFN-γ production by memory TIA cells is protective in subsequent heterologous challenge with the bacterial pathogen Legionella pneumophila. In contrast, antigen-independent reactivation of CD4+ memory TIA cells accelerates disease onset in an autoimmune model of multiple sclerosis. Our findings demonstrate that memory Th1 cells can acquire additional TCR-independent functionality to mount rapid, innate-like responses that modulate susceptibility to heterologous challenges.
Collapse
Affiliation(s)
- Nikolas Rakebrandt
- Institute of Experimental Immunology, University of Zurich, 8057Zurich, Switzerland
| | - Nima Yassini
- Institute of Experimental Immunology, University of Zurich, 8057Zurich, Switzerland
- Department of Quantitative Biomedicine, University of Zurich, 8057Zurich, Switzerland
| | - Anna Kolz
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg, Germany
| | - Michelle Schorer
- Institute of Experimental Immunology, University of Zurich, 8057Zurich, Switzerland
| | - Katharina Lambert
- Institute of Experimental Immunology, University of Zurich, 8057Zurich, Switzerland
| | - Eva Goljat
- Department of Quantitative Biomedicine, University of Zurich, 8057Zurich, Switzerland
| | - Anna Estrada Brull
- Department of Quantitative Biomedicine, University of Zurich, 8057Zurich, Switzerland
| | - Celine Rauld
- Novartis Biomedical Research, 4002Basel, Switzerland
| | - Zsolt Balazs
- Department of Quantitative Biomedicine, University of Zurich, 8057Zurich, Switzerland
| | - Michael Krauthammer
- Department of Quantitative Biomedicine, University of Zurich, 8057Zurich, Switzerland
| | | | - Anneli Peters
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg, Germany
- Biomedical Center, Faculty of Medicine, Ludwig-Maximilians-Universität München, 82152Planegg, Germany
| | - Nicole Joller
- Institute of Experimental Immunology, University of Zurich, 8057Zurich, Switzerland
- Department of Quantitative Biomedicine, University of Zurich, 8057Zurich, Switzerland
| |
Collapse
|
22
|
Wu Y, Wu Z, Jin Q, Liu J, Xu P. Identification and Analysis of Biomarkers Associated with Lipophagy and Therapeutic Agents for COVID-19. Viruses 2024; 16:923. [PMID: 38932215 PMCID: PMC11209609 DOI: 10.3390/v16060923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/28/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Lipids, as a fundamental cell component, play an regulating role in controlling the different cellular biological processes involved in viral infections. A notable feature of coronavirus disease 2019 (COVID-19) is impaired lipid metabolism. The function of lipophagy-related genes in COVID-19 is unknown. The present study aimed to investigate biomarkers and drug targets associated with lipophagy and lipophagy-based therapeutic agents for COVID-19 through bioinformatics analysis. METHODS Lipophagy-related biomarkers for COVID-19 were identified using machine learning algorithms such as random forest, Support Vector Machine-Recursive Feature Elimination, Generalized Linear Model, and Extreme Gradient Boosting in three COVID-19-associated GEO datasets: scRNA-seq (GSE145926) and bulk RNA-seq (GSE183533 and GSE190496). The cMAP database was searched for potential COVID-19 medications. RESULTS The lipophagy pathway was downregulated, and the lipid droplet formation pathway was upregulated, resulting in impaired lipid metabolism. Seven lipophagy-related genes, including ACADVL, HYOU1, DAP, AUP1, PRXAB2, LSS, and PLIN2, were used as biomarkers and drug targets for COVID-19. Moreover, lipophagy may play a role in COVID-19 pathogenesis. As prospective drugs for treating COVID-19, seven potential downregulators (phenoxybenzamine, helveticoside, lanatoside C, geldanamycin, loperamide, pioglitazone, and trichostatin A) were discovered. These medication candidates showed remarkable binding energies against the seven biomarkers. CONCLUSIONS The lipophagy-related genes ACADVL, HYOU1, DAP, AUP1, PRXAB2, LSS, and PLIN2 can be used as biomarkers and drug targets for COVID-19. Seven potential downregulators of these seven biomarkers may have therapeutic effects for treating COVID-19.
Collapse
Affiliation(s)
- Yujia Wu
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (Y.W.); (Z.W.); (Q.J.)
| | - Zhenlin Wu
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (Y.W.); (Z.W.); (Q.J.)
| | - Qiying Jin
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (Y.W.); (Z.W.); (Q.J.)
| | - Jinyuan Liu
- Basic Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China;
| | - Peiping Xu
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (Y.W.); (Z.W.); (Q.J.)
| |
Collapse
|
23
|
Xu M, Qian K, Shao H, Yao Y, Nair V, Ye J, Qin A. Metabolomics analysis of CEF cells infected with avian leukosis virus subgroup J based on UHPLC-QE-MS. Poult Sci 2024; 103:103693. [PMID: 38598912 PMCID: PMC11017069 DOI: 10.1016/j.psj.2024.103693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/21/2024] [Accepted: 03/24/2024] [Indexed: 04/12/2024] Open
Abstract
Avian leukosis virus subgroup J (ALV-J) is a retrovirus that can cause immunosuppression and tumors in chicken. However, relative pathogenesis is still not clear. At present, metabolomics has shown great potential in the screening of tumor metabolic markers, prognostic evaluation, and drug target design. In this study, we utilize an untargeted metabolomics approach based on ultrahigh-performance liquid chromatography-quadrupole time-of-flight tandem mass spectrometry (UHPLC-QTOF-MS) to analyze the metabolic changes in chicken embryo fibroblast (CEF) cells infected by ALV-J. We found that ALV-J infection significantly altered a wealth of metabolites compared with control group. Additionally, most of the differentially expressed metabolites belonged to lipid metabolism, purine nucleotide metabolism and amino acid metabolism. Among them, the proportion of lipid metabolites account for the highest proportion (around 31%). Results suggest that these changes may be conductive to the formation of virion, thereby promoting the replication of ALV-J. These data provided metabolic evidence and potential biomarkers for the cellular metabolic changes induced by ALV-J, and provided important insight for further understanding the replication needs and pathogenesis of ALV-J.
Collapse
Affiliation(s)
- Menglu Xu
- Ministry of Education Key Lab for Avian Preventive Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, P.R. China
| | - Kun Qian
- Ministry of Education Key Lab for Avian Preventive Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, P.R. China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, P.R. China
| | - Hongxia Shao
- Ministry of Education Key Lab for Avian Preventive Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, P.R. China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, P.R. China
| | - Yongxiu Yao
- The Pirbright Institute & UK-China Centre of Excellence on Avian Disease Research, Pirbright, Surrey, GU24 0NF, United Kingdom
| | - Venugopal Nair
- The Pirbright Institute & UK-China Centre of Excellence on Avian Disease Research, Pirbright, Surrey, GU24 0NF, United Kingdom
| | - Jianqiang Ye
- Ministry of Education Key Lab for Avian Preventive Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, P.R. China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, P.R. China
| | - Aijian Qin
- Ministry of Education Key Lab for Avian Preventive Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, P.R. China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, 225009, P.R. China.
| |
Collapse
|
24
|
Tang X, Zhang Y, Zhang H, Zhang N, Dai Z, Cheng Q, Li Y. Single-Cell Sequencing: High-Resolution Analysis of Cellular Heterogeneity in Autoimmune Diseases. Clin Rev Allergy Immunol 2024; 66:376-400. [PMID: 39186216 DOI: 10.1007/s12016-024-09001-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2024] [Indexed: 08/27/2024]
Abstract
Autoimmune diseases (AIDs) are complex in etiology and diverse in classification but clinically show similar symptoms such as joint pain and skin problems. As a result, the diagnosis is challenging, and usually, only broad treatments can be available. Consequently, the clinical responses in patients with different types of AIDs are unsatisfactory. Therefore, it is necessary to conduct more research to figure out the pathogenesis and therapeutic targets of AIDs. This requires research technologies with strong extraction and prediction capabilities. Single-cell sequencing technology analyses the genomic, epigenomic, or transcriptomic information at the single-cell level. It can define different cell types and states in greater detail, further revealing the molecular mechanisms that drive disease progression. These advantages enable cell biology research to achieve an unprecedented resolution and scale, bringing a whole new vision to life science research. In recent years, single-cell technology especially single-cell RNA sequencing (scRNA-seq) has been widely used in various disease research. In this paper, we present the innovations and applications of single-cell sequencing in the medical field and focus on the application contributing to the differential diagnosis and precise treatment of AIDs. Despite some limitations, single-cell sequencing has a wide range of applications in AIDs. We finally present a prospect for the development of single-cell sequencing. These ideas may provide some inspiration for subsequent research.
Collapse
Affiliation(s)
- Xuening Tang
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yudi Zhang
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Nan Zhang
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Ziyu Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Yongzhen Li
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
25
|
D’Anna SE, Vitale AM, D’Amico G, Caruso Bavisotto C, Ambrosino P, Cappello F, Maniscalco M, Marino Gammazza A. Autoimmunity against Nucleus Ambiguous Is Putatively Possible in Both Long-COVID-19 and Vaccinated Subjects: Scientific Evidence and Working Hypothesis. BIOLOGY 2024; 13:359. [PMID: 38927239 PMCID: PMC11200469 DOI: 10.3390/biology13060359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/13/2024] [Accepted: 05/18/2024] [Indexed: 06/28/2024]
Abstract
As reported by the World Health Organization (WHO), about 10-20% of people have experienced mid- to long-term effects following SARS-CoV-2 infection, collectively referred to as post-COVID-19 condition or long-COVID, including some neurovegetative symptoms. Numerous findings have suggested that the onset of these neurovegetative symptoms upon viral infection may be caused by the production of autoantibodies through molecular mimicry phenomena. Accordingly, we had previously demonstrated that 22 of the human proteins sharing putatively immunogenic peptides with SARS-CoV-2 proteins are expressed in the dorsal motor nucleus and nucleus ambiguous. Therefore, if molecular mimicry occurs following severe forms of COVID-19, there could be transitory or permanent damage in some vagal structures, resulting in a lower vagal tone and all the related clinical signs. We investigated the presence of autoantibodies against two proteins of vagal nuclei sharing a peptide with SARS-CoV-2 spike glycoprotein using an immunoassay test on blood obtained from patients with cardiorespiratory symptoms in patients affected by ongoing symptomatic COVID-19 (long-COVID), subjects vaccinated without a history of SARS-CoV-2 infection, and subjects not vaccinated without a history of SARS-CoV-2 infection. Interestingly, putative autoantibodies were present in both long-COVID-19 and vaccinated groups, opening interesting questions about pathogenic mechanisms of the disease.
Collapse
Affiliation(s)
| | - Alessandra Maria Vitale
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (A.M.V.); (G.D.); (C.C.B.); (F.C.)
| | - Giuseppa D’Amico
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (A.M.V.); (G.D.); (C.C.B.); (F.C.)
| | - Celeste Caruso Bavisotto
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (A.M.V.); (G.D.); (C.C.B.); (F.C.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Pasquale Ambrosino
- Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (S.E.D.); (P.A.)
| | - Francesco Cappello
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (A.M.V.); (G.D.); (C.C.B.); (F.C.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
- National Biodiversity Future Center (NBFC), Piazza Marina 61, 90133 Palermo, Italy
| | - Mauro Maniscalco
- Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (S.E.D.); (P.A.)
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy
| | - Antonella Marino Gammazza
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, 90127 Palermo, Italy; (A.M.V.); (G.D.); (C.C.B.); (F.C.)
| |
Collapse
|
26
|
Tsigalou C, Tsolou A, Stavropoulou E, Konstantinidis T, Zafiriou E, Dardiotis E, Tsirogianni A, Bogdanos D. Unraveling the intricate dance of the Mediterranean diet and gut microbiota in autoimmune resilience. Front Nutr 2024; 11:1383040. [PMID: 38818135 PMCID: PMC11137302 DOI: 10.3389/fnut.2024.1383040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/07/2024] [Indexed: 06/01/2024] Open
Abstract
The nutritional habits regulate the gut microbiota and increase risk of an autoimmune disease. Western diet is rich in sugars, meat, and poly-unsaturated fatty acids, which lead to dysbiosis of intestinal microbiota, disruption of gut epithelial barrier and chronic mucosal inflammation. In contrast, the Mediterranean Diet (MedDiet) is abundant in ω3 fatty acids, fruits, and vegetables, possessing anti-inflammatory properties that contribute to the restoration of gut eubiosis. Numerous studies have extensively examined the impact of MedDiet and its components on both health and various disease states. Additionally, specific investigations have explored the correlation between MedDiet, microbiota, and the risk of autoimmune diseases. Furthermore, the MedDiet has been linked to a reduced risk of cardiovascular diseases, playing a pivotal role in lowering mortality rates among individuals with autoimmune diseases and comorbidities. The aim of the present review is to specifically highlight current knowledge regarding possible interactions of MedDiet with the patterns of intestinal microbiota focusing on autoimmunity and a blueprint through dietary modulations for the prevention and management of disease's activity and progression.
Collapse
Affiliation(s)
- Christina Tsigalou
- Laboratory of Hygiene and Environmental Protection, Medical School, Democritus University of Thrace, University Hospital, Alexandroupolis, Greece
| | - Avgi Tsolou
- Laboratory of Molecular Cell Biology, Cell Cycle and Proteomics, Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - Elisavet Stavropoulou
- Laboratory of Hygiene and Environmental Protection, Medical School, Democritus University of Thrace, University Hospital, Alexandroupolis, Greece
| | - Theocharis Konstantinidis
- Laboratory of Hygiene and Environmental Protection, Medical School, Democritus University of Thrace, University Hospital, Alexandroupolis, Greece
| | - Efterpi Zafiriou
- Department of Dermatology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Efthymios Dardiotis
- Department of Neurology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Alexandra Tsirogianni
- Department of Immunology-Histocompatibility, Evangelismos General Hospital, Athens, Greece
| | - Dimitrios Bogdanos
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| |
Collapse
|
27
|
Xiao X, Cao Q, Shi Y. Thyroid tuberculosis and cold abscess after infection with COVID-19: A case report. Heliyon 2024; 10:e28469. [PMID: 38560267 PMCID: PMC10979213 DOI: 10.1016/j.heliyon.2024.e28469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 03/19/2024] [Accepted: 03/19/2024] [Indexed: 04/04/2024] Open
Abstract
There is mounting evidence that coronavirus disease 2019 (COVID-19) can cause immune dysregulation. The consequence of this immune dysregulation may contribute to susceptibility to tuberculosis (TB). Thyroid gland involvement by TB is extremely uncommon and typically the result of disseminated infection. It can be hard to diagnose because there are no identifiable symptoms. We present the case of a Chinese patient who had a fever again after COVID-19 infection that was finally diagnosed as thyroid tuberculosis with a cold abscess. Clinicians should maintain a high index of suspicion for high-risk patients from endemic regions with medical comorbidities, such as immunocompromised disease and malnutrition.
Collapse
Affiliation(s)
| | | | - Yujia Shi
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, 213003, China
| |
Collapse
|
28
|
Talukder S, Deb P, Parveen M, Zannat KE, Bhuiyan AH, Yeasmin M, Molla MMA, Saif-Ur-Rahman KM. Clinical features and outcomes of COVID-19 patients with concomitant herpesvirus co-infection or reactivation: A systematic review. New Microbes New Infect 2024; 58:101233. [PMID: 38425457 PMCID: PMC10901905 DOI: 10.1016/j.nmni.2024.101233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 02/11/2024] [Accepted: 02/14/2024] [Indexed: 03/02/2024] Open
Abstract
Background Since the first case of COVID-19 was diagnosed in Wuhan, China in late 2019, concomitant infections with Herpesviridae were documented that were presented from simple skin manifestations to severe life-threatening conditions that may lead to mortality. In this systematic review, we have included studies conducted in different parts of the world to find out the association of clinical features and outcomes of COVID-19 infection and concomitant Herpesviridae infection. Methods A comprehensive search was conducted in electronic databases including Medline through PubMed, Cochrane database, Scopus and Web of science (core collection). Two review authors independently screened the articles and extracted data. The Risk of bias assessment was done by using RoBANS tool. Results A total of 919 studies were retrieved and 19 studies were included having data of 539 patients who were infected with both COVID-19 and Herpesviridae. Herpes Simplex-1, Varicella Zoster, Cytomegalovirus, Epstein-Barr virus and Human Herpes Virus-6 were the detected viruses in the included studies. Cytomegalovirus (CMV) reactivation was the most detected concomitant infection. In case of reactivation with more than one Herpes virus mortality among patients were detected along with single viral infection in some studies. Significant association was noted in dosage and usage of steroid and Herpesviridae reactivation in COVID-19 patients. Blood markers such as D-dimer, CRP along with length of stay in the ICU and usage of invasive mechanical ventilation were found to be the significantly associated markers. Conclusion Findings from this study will aid clinicians to assess and treat COVID-19 cases with co-infections.
Collapse
Affiliation(s)
- Shiny Talukder
- Department of Microbiology, Rangamati Medical College, Rangamati, Bangladesh
| | - Paroma Deb
- Department of Microbiology and Immunology, University of Iowa, Iowa city, USA
| | - Monira Parveen
- Department of General and Dental Pharmacology, Dhaka Dental College, Dhaka, Bangladesh
| | - Kaniz E Zannat
- DMFR Molecular Laboratory and Diagnostics, Dhaka, Bangladesh
| | | | - Mahmuda Yeasmin
- Department of Virology, National Institute of Laboratory Medicine and Referral Centre, Dhaka, Bangladesh
| | - Md Maruf Ahmed Molla
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - KM Saif-Ur-Rahman
- Evidence Synthesis Ireland and Cochrane Ireland, University of Galway, Galway, Ireland
- College of Medicine, Nursing and Health Sciences, University of Galway, Galway, Ireland
| |
Collapse
|
29
|
Vasilevska V, Guest PC, Szardenings M, Benros ME, Steiner J. Possible temporal relationship between SARS-CoV-2 infection and anti-NMDA receptor encephalitis: a meta-analysis. Transl Psychiatry 2024; 14:139. [PMID: 38459000 PMCID: PMC10923949 DOI: 10.1038/s41398-024-02831-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 03/10/2024] Open
Abstract
The global impact of SARS-CoV-2 infection has raised concerns about secondary diseases beyond acute illness. This review explores the significance and potential underlying mechanisms of how SARS-CoV-2 infection might elicit an immune response targeting N-methyl-D-aspartate (NMDA) receptors, and its implications for autoimmune-driven neuropsychiatric manifestations. We identified 19 published case reports of NMDA receptor encephalitis associated with SARS-CoV-2 infection or vaccination by a systematic literature search. The significance of these reports was limited since it is not clear if a coincidental or causal relationship exists between SARS-CoV-2 infection or vaccination and manifestation of NMDA receptor encephalitis. The included studies were hampered by difficulties in establishing if these patients had pre-existing NMDA receptor antibodies which entered the brain by infection- or vaccination-associated transient blood-brain barrier leakage. In addition, four cases had comorbid ovarian teratoma, which is a known trigger for development of NMDA receptor encephalitis. Considering that billions of people have contracted COVID-19 or have been vaccinated against this virus, the publication of only 19 case reports with a possible link to NMDA receptor encephalitis, indicates that it is rare. In conclusion, these findings do not support the case that SARS-CoV-2 infection or vaccination led to an increase of existing or de novo encephalitis mediated by an autoimmune response targeting NMDA receptor function. Nevertheless, this work underscores the importance of ongoing vigilance in monitoring viral outbreaks and their potential impact on the central nervous system through basic, epidemiological and translational research.
Collapse
Affiliation(s)
- Veronika Vasilevska
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Paul C Guest
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Michael Szardenings
- Ligand Development Unit, Fraunhofer Institute of Cell Therapy and Immunology, Leipzig, Germany
| | - Michael E Benros
- Copenhagen Research Centre for Mental Health, Mental Health Center Copenhagen, Copenhagen University Hospital, Hellerup, Denmark
| | - Johann Steiner
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.
- Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany.
- Center for Health and Medical Prevention (CHaMP), Magdeburg, Germany.
- German Center for Mental Health (DZPG), Partner Site Halle-Jena-Magdeburg, Magdeburg, Germany.
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Magdeburg, Germany.
| |
Collapse
|
30
|
Bastard P, Gervais A, Le Voyer T, Philippot Q, Cobat A, Rosain J, Jouanguy E, Abel L, Zhang SY, Zhang Q, Puel A, Casanova JL. Human autoantibodies neutralizing type I IFNs: From 1981 to 2023. Immunol Rev 2024; 322:98-112. [PMID: 38193358 PMCID: PMC10950543 DOI: 10.1111/imr.13304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Human autoantibodies (auto-Abs) neutralizing type I IFNs were first discovered in a woman with disseminated shingles and were described by Ion Gresser from 1981 to 1984. They have since been found in patients with diverse conditions and are even used as a diagnostic criterion in patients with autoimmune polyendocrinopathy syndrome type 1 (APS-1). However, their apparent lack of association with viral diseases, including shingles, led to wide acceptance of the conclusion that they had no pathological consequences. This perception began to change in 2020, when they were found to underlie about 15% of cases of critical COVID-19 pneumonia. They have since been shown to underlie other severe viral diseases, including 5%, 20%, and 40% of cases of critical influenza pneumonia, critical MERS pneumonia, and West Nile virus encephalitis, respectively. They also seem to be associated with shingles in various settings. These auto-Abs are present in all age groups of the general population, but their frequency increases with age to reach at least 5% in the elderly. We estimate that at least 100 million people worldwide carry auto-Abs neutralizing type I IFNs. Here, we briefly review the history of the study of these auto-Abs, focusing particularly on their known causes and consequences.
Collapse
Affiliation(s)
- Paul Bastard
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France, EU
- Paris Cité University, Imagine Institute, Paris, France, EU
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, Assistante Publique-Hôpitaux de Paris (AP-HP), Paris, France, EU
| | - Adrian Gervais
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France, EU
- Paris Cité University, Imagine Institute, Paris, France, EU
| | - Tom Le Voyer
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France, EU
- Paris Cité University, Imagine Institute, Paris, France, EU
| | - Quentin Philippot
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France, EU
- Paris Cité University, Imagine Institute, Paris, France, EU
| | - Aurélie Cobat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France, EU
- Paris Cité University, Imagine Institute, Paris, France, EU
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Jérémie Rosain
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France, EU
- Paris Cité University, Imagine Institute, Paris, France, EU
| | - Emmanuelle Jouanguy
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France, EU
- Paris Cité University, Imagine Institute, Paris, France, EU
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France, EU
- Paris Cité University, Imagine Institute, Paris, France, EU
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Shen-Ying Zhang
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France, EU
- Paris Cité University, Imagine Institute, Paris, France, EU
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Qian Zhang
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France, EU
- Paris Cité University, Imagine Institute, Paris, France, EU
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France, EU
- Paris Cité University, Imagine Institute, Paris, France, EU
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France, EU
- Paris Cité University, Imagine Institute, Paris, France, EU
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Howard Hughes Medical Institute, New York, NY, USA
- Department of Pediatrics, Necker Hospital for Sick Children, APHP, Paris, France, EU
| |
Collapse
|
31
|
Fan L, Liang Z, Ren J, Chen Y, Zhu H, Chen Y, Xiang B, Lin Q, Ding C, Chen L, Ren T. Newcastle disease virus activates the PI3K/AKT signaling pathway by targeting PHLPP2 degradation to delay cell apoptosis and promote viral replication. Vet Microbiol 2024; 289:109949. [PMID: 38128444 DOI: 10.1016/j.vetmic.2023.109949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/27/2023] [Accepted: 12/10/2023] [Indexed: 12/23/2023]
Abstract
Newcastle disease (ND) is a highly pathogenic, contagious, and fatal infectious disease in poultry caused by the Newcastle disease virus (NDV). The PI3K/AKT signaling pathway is a phosphorylation cascade that participates in regulating several cellular functions. Viruses reportedly regulate the course of infection through the PI3K/AKT axis. Here, we aimed to analyze the pathogenesis of NDV infection mediated by the PI3K/AKT signaling pathway activation. We found that NDV infection can phosphorylate AKT to activate the PI3K/AKT axis both in vitro and in vivo. Flow cytometry and Caspase-3 activity assay showed that NDV infection could inhibit cell apoptosis. The activation or inhibition of the PI3K/AKT signaling pathway activity significantly inhibited or promoted NDV-mediated apoptosis. Furthermore, inhibition of cell apoptosis significantly promoted NDV replication. Overall, our results showed that NDV infection activates the PI3K/AKT signaling pathway and inhibits cell apoptosis, thus promoting viral replication. In this context, the reduced expression of PHLPP2 protein mediated by NDV infection could be inhibited by MG132. PHLPP2 expression reversely and positively regulated NDV replication and cell apoptosis, respectively. These results indicated that NDV infection-mediated activation of the PI3K/AKT signaling pathway and the inhibition of apoptosis depend on the ubiquitin-proteasome degradation of the PHLPP2 protein. Co-IP and indirect immunofluorescence results showed that NDV V protein could interact with PHLPP2 protein, indicating that NDV targeted PHLPP2 protein degradation through V protein to activate the PI3K/AKT signaling pathway. This study deepens our understanding of the molecular mechanisms of NDV infection, providing a theoretical basis for ND prevention and control.
Collapse
Affiliation(s)
- Lei Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, China
| | - Zhaoping Liang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, China
| | - Jinlian Ren
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, China
| | - Yichun Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, China
| | - He Zhu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, China
| | - Yanan Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, China
| | - Bin Xiang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201 Yunnan, China
| | - Qiuyan Lin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, China
| | - Chan Ding
- Shanghai Veterinary Research Institute (SHVRI), Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Libin Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, China.
| | - Tao Ren
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, China.
| |
Collapse
|
32
|
Shi X, Wallach JD, Ma X, Rogne T. Autoimmune diseases and risk of non-Hodgkin lymphoma: A Mendelian randomisation study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.20.24301459. [PMID: 38343812 PMCID: PMC10854352 DOI: 10.1101/2024.01.20.24301459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Objective To examine whether genetically predicted susceptibility to ten autoimmune diseases (Behçet's disease, coeliac disease, dermatitis herpetiformis, lupus, psoriasis, rheumatoid arthritis, sarcoidosis, Sjögren's syndrome, systemic sclerosis, and type 1 diabetes) is associated with risk of non-Hodgkin lymphoma (NHL). Design Two sample Mendelian randomization (MR) study. Setting Genome wide association studies (GWASs) of ten autoimmune diseases, NHL, and four NHL subtypes (i.e., follicular lymphoma, mature T/natural killer-cell lymphomas, non-follicular lymphoma, and other and unspecified types of NHL). Analysis We used data from the largest publicly available GWASs of European ancestry for each autoimmune disease, NHL, and NHL subtypes. For each autoimmune disease, we extracted single nucleotide polymorphisms (SNPs) strongly associated (P < 5×10-8) with that disease and that were independent of one another (R2 < 1×10-3) as genetic instruments. SNPs within the human leukocyte antigen region were not considered due to potential pleiotropy. Our primary MR analysis was the inverse-variance weighted analysis. Additionally, we conducted MR-Egger, weighted mode, and weighted median regression to address potential bias due to pleiotropy, and robust adjusted profile scores to address weak instrument bias. We carried out sensitivity analysis limited to the non-immune pathway for nominally significant findings. To account for multiple testing, we set the thresholds for statistical significance at P < 5×10-3. Participants The number of cases and controls identified in the relevant GWASs were 437 and 3,325 for Behçet's disease, 4,918 and 5,684 for coeliac disease, 435 and 341,188 for dermatitis herpetiformis, 4,576 and 8,039 for lupus, 11,988 and 275,335 for psoriasis, 22,350 and 74,823 for rheumatoid arthritis, 3,597 and 337,121 for sarcoidosis, 2,735 and 332,115 for Sjögren's syndrome, 9,095 and 17,584 for systemic sclerosis, 18,942 and 501,638 for type 1 diabetes, 2,400 and 410,350 for NHL; and 296 to 2,340 cases and 271,463 controls for NHL subtypes. Exposures Genetic variants predicting ten autoimmune diseases: Behçet's disease, coeliac disease, dermatitis herpetiformis, lupus, psoriasis, rheumatoid arthritis, sarcoidosis, Sjögren's syndrome, systemic sclerosis, and type 1 diabetes. Main outcome measures Estimated associations between genetically predicted susceptibility to ten autoimmune diseases and the risk of NHL. Results The variance of each autoimmune disease explained by the SNPs ranged from 0.3% to 3.1%. Negative associations between type 1 diabetes and sarcoidosis and the risk of NHL were observed (odds ratio [OR] 0.95, 95% confidence interval [CI]: 0.92 to 0.98, P = 5×10-3, and OR 0.92, 95% CI: 0.85 to 0.99, P = 2.8×10-2, respectively). These findings were supported by the sensitivity analyses accounting for potential pleiotropy and weak instrument bias. No significant associations were found between the other eight autoimmune diseases and NHL risk. Of the NHL subtypes, type 1 diabetes was most strongly associated with follicular lymphoma (OR 0.91, 95% CI: 0.86 to 0.96, P = 1×10-3), while sarcoidosis was most strongly associated with other and unspecified NHL (OR 0.86, 95% CI: 0.75 to 0.97, P = 1.8×10-2). Conclusions These findings suggest that genetically predicted susceptibility to type 1 diabetes, and to some extent sarcoidosis, might reduce the risk of NHL. However, future studies with different datasets, approaches, and populations are warranted to further examine the potential associations between these autoimmune diseases and the risk of NHL.
Collapse
Affiliation(s)
- Xiaoting Shi
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, Connecticut, USA
- Yale Center for Perinatal, Pediatric, and Environmental Epidemiology, Yale School of Public Health, New Haven, Connecticut, USA
| | - Joshua D. Wallach
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Xiaomei Ma
- Department of Chronic Diseases Epidemiology, Yale School of Public Health, New Haven, Connecticut, USA
| | - Tormod Rogne
- Department of Chronic Diseases Epidemiology, Yale School of Public Health, New Haven, Connecticut, USA
- Yale Center for Perinatal, Pediatric, and Environmental Epidemiology, Yale School of Public Health, New Haven, Connecticut, USA
| |
Collapse
|
33
|
Volovat SR, Scripcariu DV, Vasilache IA, Stolniceanu CR, Volovat C, Augustin IG, Volovat CC, Ostafe MR, Andreea-Voichița SG, Bejusca-Vieriu T, Lungulescu CV, Sur D, Boboc D. Oncolytic Virotherapy: A New Paradigm in Cancer Immunotherapy. Int J Mol Sci 2024; 25:1180. [PMID: 38256250 PMCID: PMC10816814 DOI: 10.3390/ijms25021180] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Oncolytic viruses (OVs) are emerging as potential treatment options for cancer. Natural and genetically engineered viruses exhibit various antitumor mechanisms. OVs act by direct cytolysis, the potentiation of the immune system through antigen release, and the activation of inflammatory responses or indirectly by interference with different types of elements in the tumor microenvironment, modification of energy metabolism in tumor cells, and antiangiogenic action. The action of OVs is pleiotropic, and they show varied interactions with the host and tumor cells. An important impediment in oncolytic virotherapy is the journey of the virus into the tumor cells and the possibility of its binding to different biological and nonbiological vectors. OVs have been demonstrated to eliminate cancer cells that are resistant to standard treatments in many clinical trials for various cancers (melanoma, lung, and hepatic); however, there are several elements of resistance to the action of viruses per se. Therefore, it is necessary to evaluate the combination of OVs with other standard treatment modalities, such as chemotherapy, immunotherapy, targeted therapies, and cellular therapies, to increase the response rate. This review provides a comprehensive update on OVs, their use in oncolytic virotherapy, and the future prospects of this therapy alongside the standard therapies currently used in cancer treatment.
Collapse
Affiliation(s)
- Simona Ruxandra Volovat
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania; (S.R.V.); (M.-R.O.); (S.-G.A.-V.); (T.B.-V.)
| | - Dragos Viorel Scripcariu
- Department of Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania;
| | - Ingrid Andrada Vasilache
- Department of Obstetrics and Gynecology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cati Raluca Stolniceanu
- Department of Biophysics and Medical Physics—Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania;
| | - Constantin Volovat
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania; (S.R.V.); (M.-R.O.); (S.-G.A.-V.); (T.B.-V.)
| | | | | | - Madalina-Raluca Ostafe
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania; (S.R.V.); (M.-R.O.); (S.-G.A.-V.); (T.B.-V.)
| | - Slevoacă-Grigore Andreea-Voichița
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania; (S.R.V.); (M.-R.O.); (S.-G.A.-V.); (T.B.-V.)
| | - Toni Bejusca-Vieriu
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania; (S.R.V.); (M.-R.O.); (S.-G.A.-V.); (T.B.-V.)
| | | | - Daniel Sur
- 11th Department of Medical Oncology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania;
| | - Diana Boboc
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania; (S.R.V.); (M.-R.O.); (S.-G.A.-V.); (T.B.-V.)
| |
Collapse
|
34
|
Boyarchuk O, Volianska L. Autoimmunity and long COVID in children. Reumatologia 2024; 61:492-501. [PMID: 38322108 PMCID: PMC10839920 DOI: 10.5114/reum/176464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 12/06/2023] [Indexed: 02/08/2024] Open
Abstract
Numerous hypotheses regarding the pathogenetic mechanisms of long COVID have been proposed. Immune dysregulation and autoimmunity are among the leading hypotheses. In this article, we present two clinical cases of long COVID. The first case demonstrates the phenotype of long COVID with pain and musculoskeletal symptoms, which is often associated with autoimmunity and mimics systemic connective tissue diseases. In the second case, a high titer of antinuclear antibodies was observed after SARS-CoV-2 infection, but the clinical symptoms were limited to fever and headache. Only a comprehensive evaluation of clinical symptoms and thorough objective examination can confirm or exclude autoimmune diseases after a previous SARS-CoV-2 infection. A systematic search in the PubMed Medline database was carried out for studies focusing on immune dysregulation, autoimmunity, and its association with the clinical phenotype of long COVID. The question of the role of autoimmunity in the development of long COVID and the management approaches are discussed.
Collapse
Affiliation(s)
- Oksana Boyarchuk
- Department of Children's Diseases and Pediatric Surgery, I. Horbachevsky Ternopil National Medical University, Ukraine
| | - Liubov Volianska
- Department of Children's Diseases and Pediatric Surgery, I. Horbachevsky Ternopil National Medical University, Ukraine
| |
Collapse
|
35
|
Dong Y, Wang T, Wu H. Tertiary lymphoid structures in autoimmune diseases. Front Immunol 2024; 14:1322035. [PMID: 38259436 PMCID: PMC10800951 DOI: 10.3389/fimmu.2023.1322035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Tertiary lymphoid structures (TLSs) are organized lymphoid-like aggregations in non-lymphoid tissues. Tissues with chronic and persistent inflammation infiltration may drive and form ectopic germinal center-like structures, which are very common in autoimmune diseases, chronic infections, and tumor microenvironments. However, the mechanisms governing the formation of TLSs are still being explored. At present, it is not clear whether the formation of TLSs is associated with local uncontrolled immune inflammatory responses. While TLSs suggest a good prognosis in tumors, the opposite is true in autoimmune diseases. This review article will discuss the current views on initiating and maintaining TLSs and the potential therapeutic target in autoimmune diseases.
Collapse
Affiliation(s)
- Yuanji Dong
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ting Wang
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Huaxiang Wu
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
36
|
He Y, Zou P, Lu J, Lu Y, Yuan S, Zheng X, Liu J, Zeng C, Liu L, Tang L, Fang Z, Hu X, Liu Q, Zhou S. CD4+ T-Cell Legumain Deficiency Attenuates Hypertensive Damage via Preservation of TRAF6. Circ Res 2024; 134:9-29. [PMID: 38047378 DOI: 10.1161/circresaha.123.322835] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 11/16/2023] [Indexed: 12/05/2023]
Abstract
BACKGROUND T cells are central to the immune responses contributing to hypertension. LGMN (legumain) is highly expressed in T cells; however, its role in the pathogenesis of hypertension remains unclear. METHODS Peripheral blood samples were collected from patients with hypertension, and cluster of differentiation (CD)4+ T cells were sorted for gene expression and Western blotting analysis. TLGMNKO (T cell-specific LGMN-knockout) mice (Lgmnf/f/CD4Cre), regulatory T cell (Treg)-specific LGMN-knockout mice (Lgmnf/f/Foxp3YFP Cre), and RR-11a (LGMN inhibitor)-treated C57BL/6 mice were infused with Ang II (angiotensin II) or deoxycorticosterone acetate/salt to establish hypertensive animal models. Flow cytometry, 4-dimensional label-free proteomics, coimmunoprecipitation, Treg suppression, and in vivo Treg depletion or adoptive transfer were used to delineate the functional importance of T-cell LGMN in hypertension development. RESULTS LGMN mRNA expression was increased in CD4+ T cells isolated from hypertensive patients and mice, was positively correlated with both systolic and diastolic blood pressure, and was negatively correlated with serum IL (interleukin)-10 levels. TLGMNKO mice exhibited reduced Ang II-induced or deoxycorticosterone acetate/salt-induced hypertension and target organ damage relative to wild-type (WT) mice. Genetic and pharmacological inhibition of LGMN blocked Ang II-induced or deoxycorticosterone acetate/salt-induced immunoinhibitory Treg reduction in the kidneys and blood. Anti-CD25 antibody depletion of Tregs abolished the protective effects against Ang II-induced hypertension in TLGMNKO mice, and LGMN deletion in Tregs prevented Ang II-induced hypertension in mice. Mechanistically, endogenous LGMN impaired Treg differentiation and function by directly interacting with and facilitating the degradation of TRAF6 (tumor necrosis factor receptor-associated factor 6) via chaperone-mediated autophagy, thereby inhibiting NF-κB (nuclear factor kappa B) activation. Adoptive transfer of LGMN-deficient Tregs reversed Ang II-induced hypertension, whereas depletion of TRAF6 in LGMN-deficient Tregs blocked the protective effects. CONCLUSIONS LGMN deficiency in T cells prevents hypertension and its complications by promoting Treg differentiation and function. Specifically targeting LGMN in Tregs may be an innovative approach for hypertension treatment.
Collapse
Affiliation(s)
- Yuhu He
- Departments of Cardiology (Y.H., P.Z., X.Z., J. Liu, C.Z., L.L., L.T., Z.F., X.H., Q.L., S.Z.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Pu Zou
- Departments of Cardiology (Y.H., P.Z., X.Z., J. Liu, C.Z., L.L., L.T., Z.F., X.H., Q.L., S.Z.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Junmi Lu
- Pathology (J. Lu), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yufei Lu
- Division of Physical Therapy Education, College of Allied Health Professions, University of Nebraska Medical Center, Omaha (Y.L.)
| | - Shuguang Yuan
- Nephrology (S.Y.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xialei Zheng
- Departments of Cardiology (Y.H., P.Z., X.Z., J. Liu, C.Z., L.L., L.T., Z.F., X.H., Q.L., S.Z.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jing Liu
- Departments of Cardiology (Y.H., P.Z., X.Z., J. Liu, C.Z., L.L., L.T., Z.F., X.H., Q.L., S.Z.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Cheng Zeng
- Departments of Cardiology (Y.H., P.Z., X.Z., J. Liu, C.Z., L.L., L.T., Z.F., X.H., Q.L., S.Z.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ling Liu
- Departments of Cardiology (Y.H., P.Z., X.Z., J. Liu, C.Z., L.L., L.T., Z.F., X.H., Q.L., S.Z.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Liang Tang
- Departments of Cardiology (Y.H., P.Z., X.Z., J. Liu, C.Z., L.L., L.T., Z.F., X.H., Q.L., S.Z.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhenfei Fang
- Departments of Cardiology (Y.H., P.Z., X.Z., J. Liu, C.Z., L.L., L.T., Z.F., X.H., Q.L., S.Z.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xinqun Hu
- Departments of Cardiology (Y.H., P.Z., X.Z., J. Liu, C.Z., L.L., L.T., Z.F., X.H., Q.L., S.Z.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Qiming Liu
- Departments of Cardiology (Y.H., P.Z., X.Z., J. Liu, C.Z., L.L., L.T., Z.F., X.H., Q.L., S.Z.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Shenghua Zhou
- Departments of Cardiology (Y.H., P.Z., X.Z., J. Liu, C.Z., L.L., L.T., Z.F., X.H., Q.L., S.Z.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
37
|
Abstract
If one had any doubts before the pandemic regarding the correlation between infections and autoimmunity, COVID-19 left us fascinated on the strong bond between the two entities. The immune and autoimmune reactions seen in patients infected with SARS-CoV-2 have served as a base for this assumption. Later on, the use of immunosuppressants such as systemic glucocorticoids, among other biological agents, turned this assumption to a fact. This was no different when it comes to the vaccines against COVID-19. Through several postulated mechanisms these vaccines, although generally considered safe, are thought to have the potential to result in autoimmune reactions making them not more innocent than the infection itself. When systemic lupus erythematous (SLE) is viewed as a classical autoimmune multisystemic disorder, the connection with SARS-CoV-2 infection and COVID-19 vaccination is of extreme importance. This is because early reports during the pandemic have shown increased rates of SARS-CoV-2 infection among patients known previously to have SLE and much more interestingly, cases of new-onset SLE after COVID-19 have been documented in the literature. Subsequently vaccines against COVID-19, those mRNA-based and adenovirus-vector based, were reported to induce new SLE cases, trigger immune thrombocytopenia or lupus nephritis, two common presentations of SLE, or exacerbate flares. In our paper, we concluded various aspects of available and recent data regarding SLE and COVID-19 as both an infection and vaccination.
Collapse
Affiliation(s)
- Naim Mahroum
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Abdulrahman Elsalti
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Mehmet Fatih Ozkan
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Ramat-Gan, Israel
- Reichman University, Herzliya, Israel
| |
Collapse
|
38
|
Mohamed MM, Ibrahim A, Razaq Z, Hassan W. A Case of Postcoronavirus Disease 2019 Antineutrophil Cytoplasmic Antibody-associated Vasculitis Successfully Treated with Rituximab. SAUDI JOURNAL OF KIDNEY DISEASES AND TRANSPLANTATION 2023; 34:S219-S225. [PMID: 38995287 DOI: 10.4103/sjkdt.sjkdt_317_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024] Open
Abstract
We report a case of a 69-year-old Caucasian male with a history of hypertension, Type 2 diabetes, and Stage IIIa chronic kidney disease (CKD), who presented to the emergency department with positional dizziness, generalized weakness, weight loss, and suppressed appetite. Two months earlier, the patient was diagnosed with coronavirus disease 2019 (COVID-19). The patient had non-oliguric acute kidney injury alongside preexisting CKD. The urinalysis showed hematuria and significant non-nephrotic proteinuria. His serological markers were positive for antineutrophil cytoplasmic antibodies with high titers. A kidney biopsy showed focal crescentic glomerulonephritis of the pauci-immune type. Initially, treatment with immunosuppressive medication was deferred because the biopsy findings suggested a poor renal outcome, as the cortical sample showed tubular atrophy and interstitial fibrosis of more than 50%. The patient was discharged but was later readmitted with worsening renal function, deep venous thrombosis in the lower extremities, and patchy lung consolidation suggesting possible pneumonia, which was ruled out. He required dialysis and brief empiric antibiotics for pneumonia, and anticoagulation for deep venous thrombosis, and was treated with intravenous (IV) pulsed steroids, followed by gradually tapering oral steroids and rituximab induction therapy. He continued dialysis three times a week. Three months after discharge, his renal function improved to near-baseline level, and he no longer required hemodialysis. He continues to be on maintenance IV rituximab therapy and low-dose oral steroids and is followed closely by a rheumatologist. Our case reflects the evolving state of understanding how COVID-19 impacts the immune system, its varying manifestations, and its management.
Collapse
Affiliation(s)
- Mahmoud M Mohamed
- Department of Internal Medicine, North Mississippi Medical Center, Tupelo, MS, USA
| | | | | | | |
Collapse
|
39
|
Zhang W, Tao Y, Zhu Y, Zheng Q, Hu F, Zhu W, Wang J, Ning M. Effect of serum autoantibodies on the COVID-19 patient's prognosis. Front Microbiol 2023; 14:1259960. [PMID: 38107861 PMCID: PMC10721969 DOI: 10.3389/fmicb.2023.1259960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/06/2023] [Indexed: 12/19/2023] Open
Abstract
Objectives Virus infection closely associated with autoimmune disease. The study aimed to explore the autoantibody profiles and the correlation of autoantibodies with the disease severity and the prognosis of the coronavirus disease 2019 (COVID-19) patients. Methods Three hundred thirty-seven hospitalized COVID-19 patients from 6th to 23rd January 2023 were enrolled. Logistic and Cox regression analyses were used to analyze the risk factors for the patient's disease severity and outcome. The association between Anti-extractable nuclear antigen antibody (ENA) positivity and the prognosis of COVID-19 patients was analyzed using Kaplan-Meier survival curves. Results 137 of COVID-19 patients were detected positive for antinuclear antibody (ANA), 61 had positive results for ENA, and 38 were positive for ANA and ENA. ANA positivity rate was higher in non-severe illness group (p = 0.032). COVID-19 patients who died during hospitalization had a high rate of ENA positivity than convalescent patients (p = 0.002). Multivariate logistic regression showed that ANA positivity was a protective factor for the disease severity of COVID-19. Multivariate Cox regression analysis revealed that ENA positivity, white blood cells count (WBC), aspartate aminotransferase (AST), Creatinine (CREA), and CRP were independent risk factors for the outcome of COVID-19 patients, and that COVID-19 patients with ENA positivity had a lower cumulative survival rate (p = 0.002). Conclusion A spectrum of autoantibodies were expressed in COVID-19 patients, among which ANA and ENA positivity was associated with the severity and prognosis of COVID-19. Therefore, autoantibodies may help to assess the disease severity and prognosis of COVID-19 patients.
Collapse
Affiliation(s)
- Weiming Zhang
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yue Tao
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yijia Zhu
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Qisi Zheng
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Fenghua Hu
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Wenbo Zhu
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Jian Wang
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Mingzhe Ning
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| |
Collapse
|
40
|
Chen K, Luo M, Lv Y, Luo Z, Yang H. Undervalued and novel roles of heterogeneous nuclear ribonucleoproteins in autoimmune diseases: Resurgence as potential biomarkers and targets. WILEY INTERDISCIPLINARY REVIEWS. RNA 2023; 14:e1806. [PMID: 37365887 DOI: 10.1002/wrna.1806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/28/2023]
Abstract
Autoimmune diseases are mainly characterized by the abnormal autoreactivity due to the loss of tolerance to specific autoantigens, though multiple pathways associated with the homeostasis of immune responses are involved in initiating or aggravating the conditions. The heterogeneous nuclear ribonucleoproteins (hnRNPs) are a major category of RNA-binding proteins ubiquitously expressed in a multitude of cells and have attracted great attentions especially with their distinctive roles in nucleic acid metabolisms and the pathogenesis in diseases like neurodegenerative disorders and cancers. Nevertheless, the interplay between hnRNPs and autoimmune disorders has not been fully elucidated. Virtually various family members of hnRNPs are increasingly identified as immune players and are pertinent to all kinds of immune-related processes including immune system development and innate or adaptive immune responses. Specifically, hnRNPs have been extensively recognized as autoantigens within and even beyond a myriad of autoimmune diseases, yet their diagnostic and prognostic values are seemingly underestimated. Molecular mimicry, epitope spreading and bystander activation may represent major putative mechanisms underlying the presence of autoantibodies to hnRNPs. Besides, hnRNPs play critical parts in regulating linchpin genes expressions that control genetic susceptibility, disease-linked functional pathways, or immune responses by interacting with other components particularly like microRNAs and long non-coding RNAs, thereby contributing to inflammation and autoimmunity as well as specific disease phenotypes. Therefore, comprehensive unraveling of the roles of hnRNPs is conducive to establishing potential biomarkers and developing better intervention strategies by targeting these hnRNPs in the corresponding disorders. This article is categorized under: RNA in Disease and Development > RNA in Disease RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications.
Collapse
Affiliation(s)
- Kangzhi Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Mengchuan Luo
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanzhi Lv
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhaohui Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center for Epileptic Disease of Hunan Province, Central South University, Changsha, China
| | - Huan Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
41
|
Hamad MN, Mohamed FI, Osman MM, Jadid AA, Abdalrhman IK, Yousif AM, Alabid T, Edris AMM, Mohamed NS, Siddig EE, Ahmed A. Molecular detection of Epstein-Barr virus among Sudanese patients diagnosed with Hashimoto's thyroiditis. BMC Res Notes 2023; 16:283. [PMID: 37858193 PMCID: PMC10588233 DOI: 10.1186/s13104-023-06399-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 06/18/2023] [Indexed: 10/21/2023] Open
Abstract
OBJECTIVES Hashimoto's thyroiditis (HT) is the most common cause of hypothyroidism. The exact mechanism initiating the development of HT is not yet clear. This study aimed to investigate the correlation between HT and the presence of Epstein-Barr virus (EBV) in a Sudanese population. RESULTS EBV-LMP1 was detected in 11.1% of HT cases, which is consistent with previous studies. Studies have reported a wide range of frequencies indicating the presence of EBV in HT, and patients with autoimmune thyroiditis have increased titers of anti-EBV antibodies in their sera compared to healthy subjects. Intrathyroidal EBV-infected B cells may be responsible for the increased risk of development of B-cell lymphoma in the thyroid gland in patients with autoimmune thyroiditis. Our study suggests that regular follow-up is necessary for patients diagnosed with HT and are positive for EBV, as antiviral therapy is not applicable due to the risk of thyroid dysfunction. The study suggests an association between EBV and HT, but causation cannot be determined. The study also highlights the need for further research to determine the viral role and correlate it with the severity and progression of HT.
Collapse
Affiliation(s)
- Marowa N Hamad
- Department of Hematology and immunology, Faculty of Medical Laboratory Sciences, University of Khartoum, Khartoum, Sudan
| | - Fuodat I Mohamed
- Department of Hematology and immunology, Faculty of Medical Laboratory Sciences, University of Khartoum, Khartoum, Sudan
| | - Mayada M Osman
- Department of Hematology and immunology, Faculty of Medical Laboratory Sciences, University of Khartoum, Khartoum, Sudan
| | - Ahlam A Jadid
- Department of Hematology and immunology, Faculty of Medical Laboratory Sciences, University of Khartoum, Khartoum, Sudan
| | - Ibtihal K Abdalrhman
- Department of Hematology and immunology, Faculty of Medical Laboratory Sciences, University of Khartoum, Khartoum, Sudan
| | - Alaa M Yousif
- Molecular Biology Unit, Sirius Training and Research Centre, Khartoum, Sudan
| | - Tyseer Alabid
- Department of Hematology and immunology, Faculty of Medical Laboratory Sciences, University of Khartoum, Khartoum, Sudan
| | - Ali Mahmoud Mohammed Edris
- Department of Histopathology and Cytology, Faculty of Medical Laboratory Sciences, University of Khartoum, Khartoum, Sudan
- Department of Histopathology and Cytology, Faculty of Applied Medical Sciences, University of Bisha, Bisha, Kingdom of Saudi Arabia
| | - Nouh S Mohamed
- Molecular Biology Unit, Sirius Training and Research Centre, Khartoum, Sudan.
| | - Emmanuel Edwar Siddig
- Faculty of Medical Laboratory Sciences, University of Khartoum, Khartoum, 11111, Sudan
- Institute of Endemic Diseases, University of Khartoum, Khartoum, 11111, Sudan
| | - Ayman Ahmed
- Institute of Endemic Diseases, University of Khartoum, Khartoum, 11111, Sudan.
- Swiss Tropical and Public Health Institute (Swiss TPH), 4123, Allschwil, Switzerland.
- University of Basel, Petersplatz 1, CH 4001, Basel, Switzerland.
| |
Collapse
|
42
|
Benoit RY, Moore CS. Epstein Barr Virus and Multiple Sclerosis: Is a Cure Possible? Viral Immunol 2023; 36:435-437. [PMID: 37724943 DOI: 10.1089/vim.2023.0109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023] Open
Affiliation(s)
- Rochelle Y Benoit
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Canada
| | - Craig S Moore
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Canada
| |
Collapse
|
43
|
Miao Y, Shi Z, Zhang W, Zhu L, Tang S, Chen H, Wang X, Du Q, Li S, Zhang Y, Luo W, Jin X, Fang M, Zhou H. Immune Repertoire Profiling Reveals Its Clinical Application Potential and Triggers for Neuromyelitis Optica Spectrum Disorders. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:e200134. [PMID: 37414573 DOI: 10.1212/nxi.0000000000200134] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 04/27/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND AND OBJECTIVES Neuromyelitis optica spectrum disorders (NMOSD) is widely recognized as a CNS demyelinating disease associated with AQP4-IgG (T cell-dependent antibody), and its trigger is still unclear. In addition, although the treatment of NMOSD currently can rely on traditional immunosuppressive and modulating agents, effective methods to predict the efficacy of these therapeutics are lacking. METHODS In this study, high-throughput T-cell receptor (TCR) sequencing was performed on peripheral blood from 151 pretreatment patients with AQP4-IgG+ NMOSD and 151 healthy individuals. We compared the TCR repertoire of those with NMOSD with that of healthy individuals and identified TCR clones that were significantly enriched in NMOSD. In addition, we treated 28 patients with AQP4-IgG+ NMOSD with immunosuppressants and followed up for 6 months to compare changes in NMOSD-specific TCRs (NMOSD-TCRs) before and after treatment. Moreover, we analyzed transcriptome and single-cell B-cell receptor (BCR) data from public databases and performed T-cell activation experiments using antigenic epitopes of cytomegalovirus (CMV) to further explore the triggers of AQP4-IgG+ NMOSD. RESULTS Compared with healthy controls, patients with AQP4-IgG+ NMOSD had significantly reduced diversity and shorter CDR3 length of TCRβ repertoire. Furthermore, we identified 597 NMOSD-TCRs with a high sequence similarity that have the potential to be used in the diagnosis and prognosis of NMOSD. The characterization of NMOSD-TCRs and pathology-associated clonotype annotation indicated that the occurrence of AQP4-IgG+ NMOSD may be associated with CMV infection, which was further corroborated by transcriptome and single-cell BCR analysis results from public databases and T-cell activation experiments. DISCUSSION Our findings suggest that the occurrence of AQP4-IgG+ NMOSD may be associated with CMV infection. In conclusion, our study provides new clues to uncover the causative factors of AQP4-IgG+ NMOSD and provides a theoretical foundation for treating and monitoring the disease.
Collapse
Affiliation(s)
- Yu Miao
- From the College of Life Sciences (M., X.J.), University of Chinese Academy of Sciences, Beijing; Department of Neurology (Z.S., L.Z., S.T., H.C., X.W., Q.D., Y.Z., W.L., M.F., H.Z.), West China Hospital, Sichuan University, Chengdu; and City University of Hong Kong (W.Z., S.L.), Shenzhen Research Institute, China
| | - Ziyan Shi
- From the College of Life Sciences (M., X.J.), University of Chinese Academy of Sciences, Beijing; Department of Neurology (Z.S., L.Z., S.T., H.C., X.W., Q.D., Y.Z., W.L., M.F., H.Z.), West China Hospital, Sichuan University, Chengdu; and City University of Hong Kong (W.Z., S.L.), Shenzhen Research Institute, China
| | - Wei Zhang
- From the College of Life Sciences (M., X.J.), University of Chinese Academy of Sciences, Beijing; Department of Neurology (Z.S., L.Z., S.T., H.C., X.W., Q.D., Y.Z., W.L., M.F., H.Z.), West China Hospital, Sichuan University, Chengdu; and City University of Hong Kong (W.Z., S.L.), Shenzhen Research Institute, China
| | - Lin Zhu
- From the College of Life Sciences (M., X.J.), University of Chinese Academy of Sciences, Beijing; Department of Neurology (Z.S., L.Z., S.T., H.C., X.W., Q.D., Y.Z., W.L., M.F., H.Z.), West China Hospital, Sichuan University, Chengdu; and City University of Hong Kong (W.Z., S.L.), Shenzhen Research Institute, China
| | - Shanshan Tang
- From the College of Life Sciences (M., X.J.), University of Chinese Academy of Sciences, Beijing; Department of Neurology (Z.S., L.Z., S.T., H.C., X.W., Q.D., Y.Z., W.L., M.F., H.Z.), West China Hospital, Sichuan University, Chengdu; and City University of Hong Kong (W.Z., S.L.), Shenzhen Research Institute, China
| | - Hongxi Chen
- From the College of Life Sciences (M., X.J.), University of Chinese Academy of Sciences, Beijing; Department of Neurology (Z.S., L.Z., S.T., H.C., X.W., Q.D., Y.Z., W.L., M.F., H.Z.), West China Hospital, Sichuan University, Chengdu; and City University of Hong Kong (W.Z., S.L.), Shenzhen Research Institute, China
| | - Xiaofei Wang
- From the College of Life Sciences (M., X.J.), University of Chinese Academy of Sciences, Beijing; Department of Neurology (Z.S., L.Z., S.T., H.C., X.W., Q.D., Y.Z., W.L., M.F., H.Z.), West China Hospital, Sichuan University, Chengdu; and City University of Hong Kong (W.Z., S.L.), Shenzhen Research Institute, China
| | - Qin Du
- From the College of Life Sciences (M., X.J.), University of Chinese Academy of Sciences, Beijing; Department of Neurology (Z.S., L.Z., S.T., H.C., X.W., Q.D., Y.Z., W.L., M.F., H.Z.), West China Hospital, Sichuan University, Chengdu; and City University of Hong Kong (W.Z., S.L.), Shenzhen Research Institute, China.
| | - Shuaicheng Li
- From the College of Life Sciences (M., X.J.), University of Chinese Academy of Sciences, Beijing; Department of Neurology (Z.S., L.Z., S.T., H.C., X.W., Q.D., Y.Z., W.L., M.F., H.Z.), West China Hospital, Sichuan University, Chengdu; and City University of Hong Kong (W.Z., S.L.), Shenzhen Research Institute, China.
| | - Ying Zhang
- From the College of Life Sciences (M., X.J.), University of Chinese Academy of Sciences, Beijing; Department of Neurology (Z.S., L.Z., S.T., H.C., X.W., Q.D., Y.Z., W.L., M.F., H.Z.), West China Hospital, Sichuan University, Chengdu; and City University of Hong Kong (W.Z., S.L.), Shenzhen Research Institute, China
| | - Wenqin Luo
- From the College of Life Sciences (M., X.J.), University of Chinese Academy of Sciences, Beijing; Department of Neurology (Z.S., L.Z., S.T., H.C., X.W., Q.D., Y.Z., W.L., M.F., H.Z.), West China Hospital, Sichuan University, Chengdu; and City University of Hong Kong (W.Z., S.L.), Shenzhen Research Institute, China
| | - Xin Jin
- From the College of Life Sciences (M., X.J.), University of Chinese Academy of Sciences, Beijing; Department of Neurology (Z.S., L.Z., S.T., H.C., X.W., Q.D., Y.Z., W.L., M.F., H.Z.), West China Hospital, Sichuan University, Chengdu; and City University of Hong Kong (W.Z., S.L.), Shenzhen Research Institute, China.
| | - Mingyan Fang
- From the College of Life Sciences (M., X.J.), University of Chinese Academy of Sciences, Beijing; Department of Neurology (Z.S., L.Z., S.T., H.C., X.W., Q.D., Y.Z., W.L., M.F., H.Z.), West China Hospital, Sichuan University, Chengdu; and City University of Hong Kong (W.Z., S.L.), Shenzhen Research Institute, China.
| | - Hongyu Zhou
- From the College of Life Sciences (M., X.J.), University of Chinese Academy of Sciences, Beijing; Department of Neurology (Z.S., L.Z., S.T., H.C., X.W., Q.D., Y.Z., W.L., M.F., H.Z.), West China Hospital, Sichuan University, Chengdu; and City University of Hong Kong (W.Z., S.L.), Shenzhen Research Institute, China.
| |
Collapse
|
44
|
Adiguzel Y, Mahroum N, Muller S, Blank M, Halpert G, Shoenfeld Y. Shared Pathogenicity Features and Sequences between EBV, SARS-CoV-2, and HLA Class I Molecule-binding Motifs with a Potential Role in Autoimmunity. Clin Rev Allergy Immunol 2023; 65:206-230. [PMID: 37505416 DOI: 10.1007/s12016-023-08962-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2023] [Indexed: 07/29/2023]
Abstract
Epstein-Barr virus (EBV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are extraordinary in their ability to activate autoimmunity as well as to induce diverse autoimmune diseases. Here we reviewed the current knowledge on their relation. Further, we suggested that molecular mimicry could be a possible common mechanism of autoimmunity induction in the susceptible individuals infected with SARS-CoV-2. Molecular mimicry between SARS-CoV-2 and human proteins, and EBV and human proteins, are present. Besides, relation of the pathogenicity associated with both coronavirus diseases and EBV supports the notion. As a proof-of-the-concept, we investigated 8mer sequences with shared 5mers of SARS-CoV-2, EBV, and human proteins, which were predicted as epitopes binding to the same human leukocyte antigen (HLA) supertype representatives. We identified significant number of human peptide sequences with predicted-affinities to the HLA-A*02:01 allele. Rest of the peptide sequences had predicted-affinities to the HLA-A*02:01, HLA-B*40:01, HLA-B*27:05, HLA-A*01:01, and HLA-B*39:01 alleles. Carriers of these serotypes can be under a higher risk of autoimmune response induction upon getting infected, through molecular mimicry-based mechanisms common to SARS-CoV-2 and EBV infections. We additionally reviewed established associations of the identified proteins with the EBV-related pathogenicity and with the autoimmune diseases.
Collapse
Affiliation(s)
- Yekbun Adiguzel
- Department of Medical Biology, School of Medicine, Atilim University, Kizilcasar Mah. 06836 Incek, Golbasi, Ankara, Turkey.
| | - Naim Mahroum
- International School of Medicine, Istanbul Medipol University, Göztepe Mah, Atatürk Cd. No:40, Beykoz, Istanbul, 34810, Turkey
| | - Sylviane Muller
- Centre National de la Recherche scientifique-Université de Strasbourg, Biotechnology and Cell Signalling Unit, Neuroimmunology and Peptide Therapeutics Team, Strasbourg Drug Discovery and Development Institute, Strasbourg, France
- University of Strasbourg Institute for Advanced Study, Strasbourg, France
- Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg, University of Strasbourg, Strasbourg, France
| | - Miri Blank
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Ramat Gan, Tel-Hashomer, 52621, Israel
| | - Gilad Halpert
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Ramat Gan, Tel-Hashomer, 52621, Israel
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Ramat Gan, Tel-Hashomer, 52621, Israel
- Reichman University, Herzliya, 4610101, Israel
| |
Collapse
|
45
|
Saeedi N, Gohari NSF, Ghalibaf AAM, Dehghan A, Owlia MB. COVID-19 infection: a possible induction factor for development of autoimmune diseases? Immunol Res 2023; 71:547-553. [PMID: 37316687 DOI: 10.1007/s12026-023-09371-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 02/14/2023] [Indexed: 06/16/2023]
Abstract
Following the global spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, the importance of investigation of the pathogenesis and immunological characteristics of COVID-19 became quite clear. Currently, there are reports indicating that COVID-19 is able to induce autoimmune responses. Abnormal immune reactions are a cornerstone in the pathogenicity of both conditions. Detection of autoantibodies in COVID-19 patients may suggest a link between COVID-19 and autoimmunity. In this study, we focused on the similarities and possible differences between COVID-19 and autoimmune disorders to explore the relationship between them. Comparing the pathogenicity of SARS-CoV-2 infection with autoimmune conditions revealed significant immunological properties of COVID-19 including the presence of several autoantibodies, autoimmunity-related cytokines, and cellular activities that could be useful in future clinical studies aiming at managing this pandemic.
Collapse
Affiliation(s)
- Nikoo Saeedi
- Student Research Committee, Islamic Azad University, Mashhad Branch, Mashhad, Iran.
| | - Narjes Sadat Farizani Gohari
- Interest Group of CoronaVirus 2019 (IGCV-19), Universal Scientific Education and Research Network (USERN), Yazd, Iran
- Student Research Committee, Faculty of Medicine, Yazd University of Medical Sciences, Yazd, Iran
| | - Amir Ali Moodi Ghalibaf
- Student Research Committee, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
- Interest Group of CoronaVirus 2019 (IGCV-19), Universal Scientific Education and Research Network (USERN), Birjand, Iran
| | - Ali Dehghan
- Division of Rheumatology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammad Bagher Owlia
- Division of Rheumatology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
46
|
Yang J, Feng J. Editorial: Viruses and immune response in pediatric infection. Front Cell Infect Microbiol 2023; 13:1257807. [PMID: 37560321 PMCID: PMC10408305 DOI: 10.3389/fcimb.2023.1257807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 08/11/2023] Open
Affiliation(s)
| | - Jiexiong Feng
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
47
|
Kervevan J, Staropoli I, Slama D, Jeger-Madiot R, Donnadieu F, Planas D, Pietri MP, Loghmari-Bouchneb W, Alaba Tanah M, Robinot R, Boufassa F, White M, Salmon-Ceron D, Chakrabarti LA. Divergent adaptive immune responses define two types of long COVID. Front Immunol 2023; 14:1221961. [PMID: 37559726 PMCID: PMC10408302 DOI: 10.3389/fimmu.2023.1221961] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 07/03/2023] [Indexed: 08/11/2023] Open
Abstract
Background The role of adaptive immune responses in long COVID remains poorly understood, with contrasting hypotheses suggesting either an insufficient antiviral response or an excessive immune response associated with inflammatory damage. To address this issue, we set to characterize humoral and CD4+ T cell responses in long COVID patients prior to SARS-CoV-2 vaccination. Methods Long COVID patients who were seropositive (LC+, n=28) or seronegative (LC-, n=23) by spike ELISA assay were recruited based on (i) an initial SARS-CoV-2 infection documented by PCR or the conjunction of three major signs of COVID-19 and (ii) the persistence or resurgence of at least 3 symptoms for over 3 months. They were compared to COVID patients with resolved symptoms (RE, n=29) and uninfected control individuals (HD, n=29). Results The spectrum of persistent symptoms proved similar in both long COVID groups, with a trend for a higher number of symptoms in the seronegative group (median=6 vs 4.5; P=0.01). The use a highly sensitive S-flow assay enabled the detection of low levels of SARS-CoV-2 spike-specific IgG in 22.7% of ELISA-seronegative long COVID (LC-) patients. In contrast, spike-specific IgG levels were uniformly high in the LC+ and RE groups. Multiplexed antibody analyses to 30 different viral antigens showed that LC- patients had defective antibody responses to all SARS-CoV-2 proteins tested but had in most cases preserved responses to other viruses. A sensitive primary T cell line assay revealed low but detectable SARS-CoV-2-specific CD4 responses in 39.1% of LC- patients, while response frequencies were high in the LC+ and RE groups. Correlation analyses showed overall strong associations between humoral and cellular responses, with exceptions in the LC- group. Conclusions These findings provide evidence for two major types of antiviral immune responses in long COVID. Seropositive patients showed coordinated cellular and humoral responses at least as high as those of recovered patients. In contrast, ELISA-seronegative long COVID patients showed overall low antiviral responses, with detectable specific CD4+ T cells and/or antibodies in close to half of patients (52.2%). These divergent findings in patients sharing a comparable spectrum of persistent symptoms raise the possibility of multiple etiologies in long COVID.
Collapse
Affiliation(s)
- Jérôme Kervevan
- Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, CNRS UMR3569, Paris, France
| | - Isabelle Staropoli
- Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, CNRS UMR3569, Paris, France
| | - Dorsaf Slama
- Department of Infectious Diseases and Immunology, Hôtel Dieu Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris Cité, Paris, France
| | - Raphaël Jeger-Madiot
- Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, CNRS UMR3569, Paris, France
| | - Françoise Donnadieu
- Infectious Disease Analytics and Epidemiology G5 Unit, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Delphine Planas
- Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, CNRS UMR3569, Paris, France
| | - Marie-Pierre Pietri
- Department of Infectious Diseases and Immunology, Hôtel Dieu Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris Cité, Paris, France
| | - Wiem Loghmari-Bouchneb
- Department of Infectious Diseases and Immunology, Hôtel Dieu Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris Cité, Paris, France
| | - Motolete Alaba Tanah
- Department of Infectious Diseases and Immunology, Hôtel Dieu Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris Cité, Paris, France
| | - Rémy Robinot
- Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, CNRS UMR3569, Paris, France
| | - Faroudy Boufassa
- INSERM U1018, Center for Research in Epidemiology and Population Health (CESP), Le Kremlin-Bicêtre, France
| | - Michael White
- Infectious Disease Analytics and Epidemiology G5 Unit, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Dominique Salmon-Ceron
- Department of Infectious Diseases and Immunology, Hôtel Dieu Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris Cité, Paris, France
| | - Lisa A. Chakrabarti
- Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, CNRS UMR3569, Paris, France
| |
Collapse
|
48
|
Gerencer M, McGuffin LJ. Are the integrin binding motifs within SARS CoV-2 spike protein and MHC class II alleles playing the key role in COVID-19? Front Immunol 2023; 14:1177691. [PMID: 37492575 PMCID: PMC10364474 DOI: 10.3389/fimmu.2023.1177691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/22/2023] [Indexed: 07/27/2023] Open
Abstract
The previous studies on the RGD motif (aa403-405) within the SARS CoV-2 spike (S) protein receptor binding domain (RBD) suggest that the RGD motif binding integrin(s) may play an important role in infection of the host cells. We also discussed the possible role of two other integrin binding motifs that are present in S protein: LDI (aa585-587) and ECD (661-663), the motifs used by some other viruses in the course of infection. The MultiFOLD models for protein structure analysis have shown that the ECD motif is clearly accessible in the S protein, whereas the RGD and LDI motifs are partially accessible. Furthermore, the amino acids that are present in Epstein-Barr virus protein (EBV) gp42 playing very important role in binding to the HLA-DRB1 molecule and in the subsequent immune response evasion, are also present in the S protein heptad repeat-2. Our MultiFOLD model analyses have shown that these amino acids are clearly accessible on the surface in each S protein chain as monomers and in the homotrimer complex and bind to HLA-DRB1 β chain. Therefore, they may have the identical role in SARS CoV-2 immune evasion as in EBV infection. The prediction analyses of the MHC class II binding peptides within the S protein have shown that the RGD motif is present in the core 9-mer peptide IRGDEVRQI within the two HLA-DRB1*03:01 and HLA-DRB3*01.01 strong binding 15-mer peptides suggesting that RGD motif may be the potential immune epitope. Accordingly, infected HLA-DRB1*03:01 or HLA-DRB3*01.01 positive individuals may develop high affinity anti-RGD motif antibodies that react with the RGD motif in the host proteins, like fibrinogen, thrombin or von Willebrand factor, affecting haemostasis or participating in autoimmune disorders.
Collapse
Affiliation(s)
| | - Liam J. McGuffin
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| |
Collapse
|
49
|
Binetti J, Real M, Renzulli M, Bertran L, Riesco D, Perpiñan C, Mohedano A, Segundo RS, Ortiz M, Porras JA, Pineda DR, Auguet T. Clinical and Biomarker Profile Responses to Rehabilitation Treatment in Patients with Long COVID Characterized by Chronic Fatigue. Viruses 2023; 15:1452. [PMID: 37515140 PMCID: PMC10384083 DOI: 10.3390/v15071452] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Long COVID (LC) syndrome is a complex multiorgan symptom that persists beyond >12 weeks after SARS-CoV-2 infection. The most frequently associated symptom is fatigue. Physical activity and exercise are recommended, although specific studies are lacking. The objectives of the present work are to analyze the impact of a supervised exercise program on the clinical evolution of LC with fatigue patients and to identify whether certain circulating biomarkers could predict the response to rehabilitation. The rehabilitation treatment response was analyzed in 14 women diagnosed with LC and fatigue, based on the changes in the 6 min walk test and Borg/Fatigue Impact scales. Patients who showed improvement in the meters walked were considered "responders" to the therapy. A total of 65% of patients responded to the exercise program, with an improvement in the meters walked and in oxygen saturation, with stability in the percentage of meters walked. Participants with obesity and those double-vaccinated against SARS-CoV-2 presented a lower degree of fatigue. LC patients presented a favorable response to a supervised exercise program. Differences in creatinine and protein levels were observed between rehabilitation therapy "responders" and "nonresponders". A good state of protein nutrition was related to a better rehabilitation response. The results are promising regarding possible predictive biomarkers of rehabilitation response, such as creatinine.
Collapse
Affiliation(s)
- Jessica Binetti
- GEMMAIR Research Group, Department of Medicine and Surgery, Pere Virgili Institute for Health Research (IISPV), Rovira i Virgili University (URV), 43007 Tarragona, Spain; (J.B.); (L.B.); (D.R.); (J.A.P.)
- Internal Medicine Unit, Joan XXIII University Hospital of Tarragona, 43007 Tarragona, Spain; (M.R.); (A.M.)
| | - Monica Real
- Internal Medicine Unit, Joan XXIII University Hospital of Tarragona, 43007 Tarragona, Spain; (M.R.); (A.M.)
| | - Marcela Renzulli
- Rehabilitation Unit, Joan XXIII University Hospital of Tarragona, 43007 Tarragona, Spain; (M.R.); (R.S.S.); (M.O.); (D.R.P.)
| | - Laia Bertran
- GEMMAIR Research Group, Department of Medicine and Surgery, Pere Virgili Institute for Health Research (IISPV), Rovira i Virgili University (URV), 43007 Tarragona, Spain; (J.B.); (L.B.); (D.R.); (J.A.P.)
| | - David Riesco
- GEMMAIR Research Group, Department of Medicine and Surgery, Pere Virgili Institute for Health Research (IISPV), Rovira i Virgili University (URV), 43007 Tarragona, Spain; (J.B.); (L.B.); (D.R.); (J.A.P.)
- Internal Medicine Unit, Joan XXIII University Hospital of Tarragona, 43007 Tarragona, Spain; (M.R.); (A.M.)
| | | | - Alba Mohedano
- Internal Medicine Unit, Joan XXIII University Hospital of Tarragona, 43007 Tarragona, Spain; (M.R.); (A.M.)
| | - Rosa San Segundo
- Rehabilitation Unit, Joan XXIII University Hospital of Tarragona, 43007 Tarragona, Spain; (M.R.); (R.S.S.); (M.O.); (D.R.P.)
- Neurobehavioral and Health Research Group (NEUROLAB), Rovira i Virgili University (URV), 43007 Tarragona, Spain
| | - Marta Ortiz
- Rehabilitation Unit, Joan XXIII University Hospital of Tarragona, 43007 Tarragona, Spain; (M.R.); (R.S.S.); (M.O.); (D.R.P.)
| | - José Antonio Porras
- GEMMAIR Research Group, Department of Medicine and Surgery, Pere Virgili Institute for Health Research (IISPV), Rovira i Virgili University (URV), 43007 Tarragona, Spain; (J.B.); (L.B.); (D.R.); (J.A.P.)
- Internal Medicine Unit, Joan XXIII University Hospital of Tarragona, 43007 Tarragona, Spain; (M.R.); (A.M.)
| | - Daniela Rosanna Pineda
- Rehabilitation Unit, Joan XXIII University Hospital of Tarragona, 43007 Tarragona, Spain; (M.R.); (R.S.S.); (M.O.); (D.R.P.)
| | - Teresa Auguet
- GEMMAIR Research Group, Department of Medicine and Surgery, Pere Virgili Institute for Health Research (IISPV), Rovira i Virgili University (URV), 43007 Tarragona, Spain; (J.B.); (L.B.); (D.R.); (J.A.P.)
- Internal Medicine Unit, Joan XXIII University Hospital of Tarragona, 43007 Tarragona, Spain; (M.R.); (A.M.)
| |
Collapse
|
50
|
Muthye V, Wasmuth JD. Proteome-wide comparison of tertiary protein structures reveals molecular mimicry in Plasmodium-human interactions. FRONTIERS IN PARASITOLOGY 2023; 2:1162697. [PMID: 39816809 PMCID: PMC11732093 DOI: 10.3389/fpara.2023.1162697] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/05/2023] [Indexed: 01/18/2025]
Abstract
Introduction Molecular mimicry is a strategy used by parasites to evade the host's immune system and facilitate transmission to a new host. To date, high-throughput examples of molecular mimicry have been limited to comparing protein sequences. However, recent advances in the prediction of tertiary structural models, led by Deepmind's AlphaFold, enable the comparison of thousands of proteins from parasites and their hosts at the structural level, allowing for the identification of more mimics. Here, we present the first proteome-level search for tertiary structure similarity between proteins from Plasmodium falciparum, a malaria-causing parasite, and humans. Methods We assembled a database of experimentally-characterized protein tertiary structures (from the Protein Data Bank) and AlphaFold-generated protein tertiary structures from P. falciparum, human, and 15 negative control species, i.e., species not infected by P. falciparum. We aligned human and control structures to the parasite structures using Foldseek. Results We identified molecular mimicry in three proteins that have been previously proposed as mediators of Plasmodium-human interactions. By extending this approach to all P. falciparum proteins, we identified an additional 41 potential mimics that are supported by additional experimental data. Discussion Our findings demonstrate a valuable application of AlphaFold-derived tertiary structural models, and we discuss key considerations for its effective use in other host-parasite systems.
Collapse
Affiliation(s)
- Viraj Muthye
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Host-Parasite Interactions Research Training Network, University of Calgary, Calgary, AB, Canada
| | - James D. Wasmuth
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Host-Parasite Interactions Research Training Network, University of Calgary, Calgary, AB, Canada
| |
Collapse
|