1
|
Huang YM, Shih LJ, Hsieh TW, Tsai KW, Lu KC, Liao MT, Hu WC. Type 2 hypersensitivity disorders, including systemic lupus erythematosus, Sjögren's syndrome, Graves' disease, myasthenia gravis, immune thrombocytopenia, autoimmune hemolytic anemia, dermatomyositis, and graft-versus-host disease, are THαβ-dominant autoimmune diseases. Virulence 2024; 15:2404225. [PMID: 39267271 PMCID: PMC11409508 DOI: 10.1080/21505594.2024.2404225] [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: 02/29/2024] [Revised: 08/21/2024] [Accepted: 09/05/2024] [Indexed: 09/17/2024] Open
Abstract
The THαβ host immunological pathway contributes to the response to infectious particles (viruses and prions). Furthermore, there is increasing evidence for associations between autoimmune diseases, and particularly type 2 hypersensitivity disorders, and the THαβ immune response. For example, patients with systemic lupus erythematosus often produce anti-double stranded DNA antibodies and anti-nuclear antibodies and show elevated levels of type 1 interferons, type 3 interferons, interleukin-10, IgG1, and IgA1 throughout the disease course. These cytokines and antibody isotypes are associated with the THαβ host immunological pathway. Similarly, the type 2 hypersensitivity disorders myasthenia gravis, Graves' disease, graft-versus-host disease, autoimmune hemolytic anemia, immune thrombocytopenia, dermatomyositis, and Sjögren's syndrome have also been linked to the THαβ pathway. Considering the potential associations between these diseases and dysregulated THαβ immune responses, therapeutic strategies such as anti-interleukin-10 or anti-interferon α/β could be explored for effective management.
Collapse
Affiliation(s)
- Yao-Ming Huang
- Department of Emergency Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
| | - Li-Jane Shih
- Department of Medical Laboratory, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
- Graduate Institute of Medical Science, National Defense Medical Center, Taipei city, Taiwan
| | - Teng-Wei Hsieh
- Division of Immunology, Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Kuo-Wang Tsai
- Department of Medical Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Kuo-Cheng Lu
- Division of Nephrology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Min-Tser Liao
- Department of Pediatrics, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
| | - Wan-Chung Hu
- Department of Clinical Pathology, Taipei Tzu Chi Hospital, Buddhist Medical Tzu Chi Foundation, New Taipei City, Taiwan
- Department of Biotechnology, Ming Chuan University, Taoyuan, Taiwan
| |
Collapse
|
2
|
Nicholas CA, Tensun FA, Evans SA, Toole KP, Broncucia H, Hesselberth JR, Gottlieb PA, Wells KL, Smith MJ. Islet-antigen reactive B cells display a unique phenotype and BCR repertoire in autoantibody positive and recent-onset type 1 diabetes patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.20.599914. [PMID: 38979376 PMCID: PMC11230262 DOI: 10.1101/2024.06.20.599914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Autoreactive B cells play an important but ill-defined role in autoimmune type 1 diabetes (T1D). To better understand their contribution, we performed single cell gene and BCR-seq analysis on pancreatic islet antigen-reactive (IAR) B cells from the peripheral blood of nondiabetic (ND), autoantibody positive prediabetic (AAB), and recent-onset T1D individuals. We found that the frequency of IAR B cells was increased in AAB and T1D. IAR B cells from these donors had altered expression of B cell signaling, pro-inflammatory, infection, and antigen processing and presentation genes. Both AAB and T1D donors demonstrated a significant increase in certain heavy and light chain V genes, and these V genes were enriched in islet-reactivity. Public clones of IAR B cells were restricted almost entirely to AAB and T1D donors. IAR B cells were clonally expanded in the autoimmune donors, particularly the AAB group. Notably, a substantial fraction of IAR B cells in AAB and T1D donors appeared to be polyreactive, which was corroborated by analysis of recombinant monoclonal antibodies. These results expand our understanding of autoreactive B cell activation during T1D and identify unique BCR repertoire changes that may serve as biomarkers for increased disease risk.
Collapse
Affiliation(s)
- Catherine A. Nicholas
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO 80045, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80045, USA
- Molecular Biology Graduate Program, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Fatima A. Tensun
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Spencer A. Evans
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Kevin P. Toole
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Hali Broncucia
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Jay R Hesselberth
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80045, USA
- Molecular Biology Graduate Program, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Peter A. Gottlieb
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO 80045, USA
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Kristen L. Wells
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO 80045, USA
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Mia J. Smith
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO 80045, USA
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| |
Collapse
|
3
|
Tóth E, Fagyas M, Nagy B, Siket IM, Szőke B, Mártha L, Mahdi M, Erdősi G, Pólik Z, Kappelmayer J, Papp Z, Borbély A, Szabó T, Balla J, Balla G, Bácsi A, Szekanecz Z, Bai P, Tóth A. Distinct subsets of anti-pulmonary autoantibodies correlate with disease severity and survival in severe COVID-19 patients. GeroScience 2024; 46:1561-1574. [PMID: 37656328 PMCID: PMC10828368 DOI: 10.1007/s11357-023-00887-2] [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: 04/25/2023] [Accepted: 07/24/2023] [Indexed: 09/02/2023] Open
Abstract
Autoantibodies targeting the lung tissue were identified in severe COVID-19 patients in this retrospective study. Fifty-three percent of 104 patients developed anti-pulmonary antibodies, the majority of which were IgM class, suggesting that they developed upon infection with SARS-CoV-2. Anti-pulmonary antibodies correlated with worse pulmonary function and a higher risk of multiorgan failure that was further aggravated if 3 or more autoantibody clones were simultaneously present (multi-producers). Multi-producer patients were older than the patients with less or no autoantibodies. One of the identified autoantibodies (targeting a pulmonary protein of ~ 50 kDa) associated with worse clinical outcomes, including mortality. In summary, severe COVID-19 is associated with the development of lung-specific autoantibodies, which may worsen the clinical outcome. Tissue proteome-wide tests, such as the ones applied here, can be used to detect autoimmunity in the post-COVID state to identify the cause of symptoms and to reveal a new target for treatment.
Collapse
Affiliation(s)
- Emese Tóth
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary
- Center of Excellence, The Hungarian Academy of Sciences, Budapest, Hungary
| | - Miklós Fagyas
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Béla Nagy
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ivetta Mányiné Siket
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Blanka Szőke
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Lilla Mártha
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Mohamed Mahdi
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gábor Erdősi
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsófia Pólik
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - János Kappelmayer
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Budapest, Hungary
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary
| | - Attila Borbély
- Division of Cardiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Szabó
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - József Balla
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Budapest, Hungary
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - György Balla
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Budapest, Hungary
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Bácsi
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Szekanecz
- Department of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary.
- Center of Excellence, The Hungarian Academy of Sciences, Budapest, Hungary.
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary.
- MTA-DE Cell Biology and Signaling Research Group ELKH, Debrecen, 4032, Hungary.
- MTA-DE Lendület Laboratory of Cellular Metabolism, 4032, Debrecen, Hungary.
| | - Attila Tóth
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, Budapest, Hungary.
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary.
| |
Collapse
|
4
|
Perera DR, Ranadeva ND, Sirisena K, Wijesinghe KJ. Roles of NS1 Protein in Flavivirus Pathogenesis. ACS Infect Dis 2024; 10:20-56. [PMID: 38110348 DOI: 10.1021/acsinfecdis.3c00566] [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] [Indexed: 12/20/2023]
Abstract
Flaviviruses such as dengue, Zika, and West Nile viruses are highly concerning pathogens that pose significant risks to public health. The NS1 protein is conserved among flaviviruses and is synthesized as a part of the flavivirus polyprotein. It plays a critical role in viral replication, disease progression, and immune evasion. Post-translational modifications influence NS1's stability, secretion, antigenicity, and interactions with host factors. NS1 protein forms extensive interactions with host cellular proteins allowing it to affect vital processes such as RNA processing, gene expression regulation, and cellular homeostasis, which in turn influence viral replication, disease pathogenesis, and immune responses. NS1 acts as an immune evasion factor by delaying complement-dependent lysis of infected cells and contributes to disease pathogenesis by inducing endothelial cell damage and vascular leakage and triggering autoimmune responses. Anti-NS1 antibodies have been shown to cross-react with host endothelial cells and platelets, causing autoimmune destruction that is hypothesized to contribute to disease pathogenesis. However, in contrast, immunization of animal models with the NS1 protein confers protection against lethal challenges from flaviviruses such as dengue and Zika viruses. Understanding the multifaceted roles of NS1 in flavivirus pathogenesis is crucial for effective disease management and control. Therefore, further research into NS1 biology, including its host protein interactions and additional roles in disease pathology, is imperative for the development of strategies and therapeutics to combat flavivirus infections successfully. This Review provides an in-depth exploration of the current available knowledge on the multifaceted roles of the NS1 protein in the pathogenesis of flaviviruses.
Collapse
Affiliation(s)
- Dayangi R Perera
- Department of Chemistry, Faculty of Science, University of Colombo, Sri Lanka 00300
| | - Nadeeka D Ranadeva
- Department of Biomedical Science, Faculty of Health Sciences, KIU Campus Sri Lanka 10120
| | - Kavish Sirisena
- Department of Chemistry, Faculty of Science, University of Colombo, Sri Lanka 00300
- Section of Genetics, Institute for Research and Development in Health and Social Care, Sri Lanka 10120
| | | |
Collapse
|
5
|
Ward KE, Steadman L, Karim AR, Reynolds GM, Pugh M, Chua W, Faustini SE, Veenith T, Thwaites RS, Openshaw PJM, Drayson MT, Shields AM, Cunningham AF, Wraith DC, Richter AG. SARS-CoV-2 infection is associated with anti-desmoglein 2 autoantibody detection. Clin Exp Immunol 2023; 213:243-251. [PMID: 37095599 PMCID: PMC10651225 DOI: 10.1093/cei/uxad046] [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: 08/23/2022] [Revised: 03/08/2023] [Accepted: 04/24/2023] [Indexed: 04/26/2023] Open
Abstract
Post-acute cardiac sequelae, following SARS-CoV-2 infection, are well recognized as complications of COVID-19. We have previously shown the persistence of autoantibodies against antigens in skin, muscle, and heart in individuals following severe COVID-19; the most common staining on skin tissue displayed an inter-cellular cement pattern consistent with antibodies against desmosomal proteins. Desmosomes play a critical role in maintaining the structural integrity of tissues. For this reason, we analyzed desmosomal protein levels and the presence of anti-desmoglein (DSG) 1, 2, and 3 antibodies in acute and convalescent sera from patients with COVID-19 of differing clinical severity. We find increased levels of DSG2 protein in sera from acute COVID-19 patients. Furthermore, we find that DSG2 autoantibody levels are increased significantly in convalescent sera following severe COVID-19 but not in hospitalized patients recovering from influenza infection or healthy controls. Levels of autoantibody in sera from patients with severe COVID-19 were comparable to levels in patients with non-COVID-19-associated cardiac disease, potentially identifying DSG2 autoantibodies as a novel biomarker for cardiac damage. To determine if there was any association between severe COVID-19 and DSG2, we stained post-mortem cardiac tissue from patients who died from COVID-19 infection. This confirmed DSG2 protein within the intercalated discs and disruption of the intercalated disc between cardiomyocytes in patients who died from COVID-19. Our results reveal the potential for DSG2 protein and autoimmunity to DSG2 to contribute to unexpected pathologies associated with COVID-19 infection.
Collapse
Affiliation(s)
- Kerensa E Ward
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Lora Steadman
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Abid R Karim
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Gary M Reynolds
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Matthew Pugh
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, West Midlands, UK
| | - Winnie Chua
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, West Midlands, UK
| | - Sian E Faustini
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Tonny Veenith
- Department of Critical Care, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Mark T Drayson
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Adrian M Shields
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
- Department of Clinical Immunology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Adam F Cunningham
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - David C Wraith
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Alex G Richter
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
- Department of Clinical Immunology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
6
|
Needham EJ, Ren AL, Digby RJ, Norton EJ, Ebrahimi S, Outtrim JG, Chatfield DA, Manktelow AE, Leibowitz MM, Newcombe VFJ, Doffinger R, Barcenas-Morales G, Fonseca C, Taussig MJ, Burnstein RM, Samanta RJ, Dunai C, Sithole N, Ashton NJ, Zetterberg H, Gisslén M, Edén A, Marklund E, Openshaw PJM, Dunning J, Griffiths MJ, Cavanagh J, Breen G, Irani SR, Elmer A, Kingston N, Summers C, Bradley JR, Taams LS, Michael BD, Bullmore ET, Smith KGC, Lyons PA, Coles AJ, Menon DK. Brain injury in COVID-19 is associated with dysregulated innate and adaptive immune responses. Brain 2022; 145:4097-4107. [PMID: 36065116 PMCID: PMC9494359 DOI: 10.1093/brain/awac321] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/24/2022] [Accepted: 08/01/2022] [Indexed: 11/30/2022] Open
Abstract
COVID-19 is associated with neurological complications including stroke, delirium and encephalitis. Furthermore, a post-viral syndrome dominated by neuropsychiatric symptoms is common, and is seemingly unrelated to COVID-19 severity. The true frequency and underlying mechanisms of neurological injury are unknown, but exaggerated host inflammatory responses appear to be a key driver of COVID-19 severity. We investigated the dynamics of, and relationship between, serum markers of brain injury [neurofilament light (NfL), glial fibrillary acidic protein (GFAP) and total tau] and markers of dysregulated host response (autoantibody production and cytokine profiles) in 175 patients admitted with COVID-19 and 45 patients with influenza. During hospitalization, sera from patients with COVID-19 demonstrated elevations of NfL and GFAP in a severity-dependent manner, with evidence of ongoing active brain injury at follow-up 4 months later. These biomarkers were associated with elevations of pro-inflammatory cytokines and the presence of autoantibodies to a large number of different antigens. Autoantibodies were commonly seen against lung surfactant proteins but also brain proteins such as myelin associated glycoprotein. Commensurate findings were seen in the influenza cohort. A distinct process characterized by elevation of serum total tau was seen in patients at follow-up, which appeared to be independent of initial disease severity and was not associated with dysregulated immune responses unlike NfL and GFAP. These results demonstrate that brain injury is a common consequence of both COVID-19 and influenza, and is therefore likely to be a feature of severe viral infection more broadly. The brain injury occurs in the context of dysregulation of both innate and adaptive immune responses, with no single pathogenic mechanism clearly responsible.
Collapse
Affiliation(s)
- Edward J Needham
- Correspondence to: Edward Needham Department of Clinical Neurosciences University of Cambridge, Cambridge, UK E-mail:
| | - Alexander L Ren
- Division of Anaesthesia, Department of Medicine, University of Cambridge, UK
| | - Richard J Digby
- Division of Anaesthesia, Department of Medicine, University of Cambridge, UK
| | - Emma J Norton
- Division of Anaesthesia, Department of Medicine, University of Cambridge, UK
| | - Soraya Ebrahimi
- Division of Anaesthesia, Department of Medicine, University of Cambridge, UK
| | - Joanne G Outtrim
- Division of Anaesthesia, Department of Medicine, University of Cambridge, UK
| | - Doris A Chatfield
- Division of Anaesthesia, Department of Medicine, University of Cambridge, UK
| | - Anne E Manktelow
- Division of Anaesthesia, Department of Medicine, University of Cambridge, UK
| | - Maya M Leibowitz
- Division of Anaesthesia, Department of Medicine, University of Cambridge, UK
| | | | - Rainer Doffinger
- Department of Clinical Biochemistry and Immunology, Addenbrooke’s Hospital, Cambridge, UK
| | | | - Claudia Fonseca
- Cambridge Protein Arrays Ltd, Babraham Research Campus, Cambridge, UK
| | - Michael J Taussig
- Cambridge Protein Arrays Ltd, Babraham Research Campus, Cambridge, UK
| | - Rowan M Burnstein
- Division of Anaesthesia, Department of Medicine, University of Cambridge, UK
| | - Romit J Samanta
- Division of Anaesthesia, Department of Medicine, University of Cambridge, UK
| | - Cordelia Dunai
- Clinical Infection Microbiology and Neuroimmunology, Institute of Infection, Veterinary and Ecological Science, Liverpool, UK
| | - Nyarie Sithole
- Department of Infectious Diseases, Cambridge University NHS Hospitals Foundation Trust, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
- Jeffrey Cheah Biomedical Centre, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Magnus Gisslén
- Department of Infectious Diseases, Institute of Biomedicine, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Infectious Diseases, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Arden Edén
- Department of Infectious Diseases, Institute of Biomedicine, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Infectious Diseases, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Emelie Marklund
- Department of Infectious Diseases, Institute of Biomedicine, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Infectious Diseases, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | - Jake Dunning
- Nuffield Department of Medicine, Pandemic Sciences Institute, University of Oxford, Oxford, UK
| | - Michael J Griffiths
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Jonathan Cavanagh
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Gerome Breen
- Department of Social Genetic and Developmental Psychiatry, King’s College London, London, UK
| | - Sarosh R Irani
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Department of Neurology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Anne Elmer
- Cambridge Clinical Research Centre, NIHR Clinical Research Facility, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge, UK
| | - Nathalie Kingston
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
- Department of Haematology, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Charlotte Summers
- Division of Anaesthesia, Department of Medicine, University of Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - John R Bradley
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
| | - Leonie S Taams
- Centre for Inflammation Biology and Cancer Immunology (CIBCI) and Department Inflammation Biology, School of Immunology and Microbial Sciences, King’s College London, Guy's Campus, London, UK
| | - Benedict D Michael
- Clinical Infection Microbiology and Neuroimmunology, Institute of Infection, Veterinary and Ecological Science, Liverpool, UK
| | - Edward T Bullmore
- Department of Psychiatry, University of Cambridge, Herchel Smith Building for Brain and Mind Sciences, Cambridge Biomedical Campus, Cambridge, UK
| | - Kenneth G C Smith
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
- Jeffrey Cheah Biomedical Centre, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
| | - Paul A Lyons
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
- Jeffrey Cheah Biomedical Centre, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
| | - Alasdair J Coles
- Department of Clinical Neurosciences, University of Cambridge, UK
| | - David K Menon
- Division of Anaesthesia, Department of Medicine, University of Cambridge, UK
| | | | | | | |
Collapse
|
7
|
Taeschler P, Cervia C, Zurbuchen Y, Hasler S, Pou C, Tan Z, Adamo S, Raeber ME, Bächli E, Rudiger A, Stüssi‐Helbling M, Huber LC, Brodin P, Nilsson J, Probst‐Müller E, Boyman O. Autoantibodies in COVID-19 correlate with antiviral humoral responses and distinct immune signatures. Allergy 2022; 77:2415-2430. [PMID: 35364615 PMCID: PMC9111424 DOI: 10.1111/all.15302] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/08/2022] [Accepted: 03/20/2022] [Indexed: 01/08/2023]
Abstract
BACKGROUND Several autoimmune features occur during coronavirus disease 2019 (COVID-19), with possible implications for disease course, immunity, and autoimmune pathology. In this study, we longitudinally screened for clinically relevant systemic autoantibodies to assess their prevalence, temporal trajectory, and association with immunity, comorbidities, and severity of COVID-19. METHODS We performed highly sensitive indirect immunofluorescence assays to detect antinuclear antibodies (ANA) and antineutrophil cytoplasmic antibodies (ANCA), along with serum proteomics and virome-wide serological profiling in a multicentric cohort of 175 COVID-19 patients followed up to 1 year after infection, eleven vaccinated individuals, and 41 unexposed controls. RESULTS Compared with healthy controls, similar prevalence and patterns of ANA were present in patients during acute COVID-19 and recovery. However, the paired analysis revealed a subgroup of patients with transient presence of certain ANA patterns during acute COVID-19. Furthermore, patients with severe COVID-19 exhibited a high prevalence of ANCA during acute disease. These autoantibodies were quantitatively associated with higher SARS-CoV-2-specific antibody titers in COVID-19 patients and in vaccinated individuals, thus linking autoantibody production to increased antigen-specific humoral responses. Notably, the qualitative breadth of antibodies cross-reactive with other coronaviruses was comparable in ANA-positive and ANA-negative individuals during acute COVID-19. In autoantibody-positive patients, multiparametric characterization demonstrated an inflammatory signature during acute COVID-19 and alterations of the B-cell compartment after recovery. CONCLUSION Highly sensitive indirect immunofluorescence assays revealed transient autoantibody production during acute SARS-CoV-2 infection, while the presence of autoantibodies in COVID-19 patients correlated with increased antiviral humoral immune responses and inflammatory immune signatures.
Collapse
Affiliation(s)
| | - Carlo Cervia
- Department of ImmunologyUniversity Hospital ZurichZurichSwitzerland
| | - Yves Zurbuchen
- Department of ImmunologyUniversity Hospital ZurichZurichSwitzerland
| | - Sara Hasler
- Department of ImmunologyUniversity Hospital ZurichZurichSwitzerland
| | - Christian Pou
- Science for Life LaboratoryDepartment of Women's and Children's HealthKarolinska InstitutetSolnaSweden
| | - Ziyang Tan
- Science for Life LaboratoryDepartment of Women's and Children's HealthKarolinska InstitutetSolnaSweden
| | - Sarah Adamo
- Department of ImmunologyUniversity Hospital ZurichZurichSwitzerland
| | - Miro E. Raeber
- Department of ImmunologyUniversity Hospital ZurichZurichSwitzerland
| | - Esther Bächli
- Clinic for Internal MedicineHirslanden Klinik St. AnnaLucerneSwitzerland
| | - Alain Rudiger
- Department of MedicineLimmattal HospitalSchlierenSwitzerland
| | | | - Lars C. Huber
- Clinic for Internal MedicineCity Hospital Triemli ZurichZurichSwitzerland
| | - Petter Brodin
- Science for Life LaboratoryDepartment of Women's and Children's HealthKarolinska InstitutetSolnaSweden
- Pediatric RheumatologyKarolinska University HospitalSolnaSweden
- Department of Immunology and InflammationImperial College LondonLondonUK
| | - Jakob Nilsson
- Department of ImmunologyUniversity Hospital ZurichZurichSwitzerland
| | | | - Onur Boyman
- Department of ImmunologyUniversity Hospital ZurichZurichSwitzerland
- Faculty of MedicineUniversity of ZurichZurichSwitzerland
| |
Collapse
|
8
|
Qi H, Liu B, Wang X, Zhang L. The humoral response and antibodies against SARS-CoV-2 infection. Nat Immunol 2022; 23:1008-1020. [PMID: 35761083 DOI: 10.1038/s41590-022-01248-5] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/22/2022] [Indexed: 12/14/2022]
Abstract
Two and a half years into the COVID-19 pandemic, we have gained many insights into the human antibody response to the causative SARS-CoV-2 virus. In this Review, we summarize key observations of humoral immune responses in people with COVID-19, discuss key features of infection- and vaccine-induced neutralizing antibodies, and consider vaccine designs for inducing antibodies that are broadly protective against different variants of the SARS-CoV-2 virus.
Collapse
Affiliation(s)
- Hai Qi
- Tsinghua-Peking Center for Life Sciences, Beijing, China. .,Laboratory of Dynamic Immunobiology, Institute for Immunology, Tsinghua University, Beijing, China. .,Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China. .,NexVac Research Center, Tsinghua University, Beijing, China. .,Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China. .,Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China.
| | - Bo Liu
- Tsinghua-Peking Center for Life Sciences, Beijing, China.,Laboratory of Dynamic Immunobiology, Institute for Immunology, Tsinghua University, Beijing, China.,Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Xinquan Wang
- Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China
| | - Linqi Zhang
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China. .,NexVac Research Center, Tsinghua University, Beijing, China. .,Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China. .,Comprehensive AIDS Research Center, Center for Global Health and Infectious Diseases, Tsinghua University, Beijing, China.
| |
Collapse
|
9
|
Song Y, Zhang Y, Li Z, Liu J, Xiao J, Song H. Potential risk factors for the development from immune thrombocytopenia to systemic lupus erythematosus: a case-control study in Chinese children. Ann Hematol 2022; 101:1447-1456. [PMID: 35532821 DOI: 10.1007/s00277-022-04836-5] [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: 12/20/2021] [Accepted: 04/06/2022] [Indexed: 11/26/2022]
Abstract
Immune thrombocytopenia (ITP) patients are at risk developing to systemic lupus erythematosus (SLE) in the future. Our study attempted to explore the potential risk factors for the development from ITP to SLE in Chinese children by statistical analysis. This study was a retrospective case-control study. Patients diagnosed with ITP and developed to SLE after the diagnosis of ITP were defined as the case group. The control group consisted of children with ITP but without developing to SLE was recruited with a ratio of 1:2. Besides univariable analysis, multivariable logistic regression was built to evaluate the potential risk factors. A total of 150 children was included with 50 in the case group and 100 in the control group. Median developing time from ITP to SLE was 34.5 [IQR 12.5, 58.75] months. ANA was found significantly different between the two groups in our study in the univariable analysis but not in the multivariable analysis (OR = 4.50, 95% CI 0.97 to 21.01). Age diagnosed ITP was positively associated with SLE (OR = 1.07 every 5 years, 95% CI 1.01 to 1.15) with alert point at 8 years old (sensitivity 0.82, specificity 0.60). A lower level of complement was also positively associated with SLE (OR = 8.33, 95% CI 1.62 to 42.91). A minimum 3-year of close follow-up for pediatric ITP patients was recommended to monitor the risk for developing SLE. Older age and hypocomplementemia were potential risk factors for the development from ITP to SLE.
Collapse
Affiliation(s)
- Yuqing Song
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 ShuaiFuYuan Street, Dongcheng District, Beijing, 100730, China
| | - Yuelun Zhang
- Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhuo Li
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 ShuaiFuYuan Street, Dongcheng District, Beijing, 100730, China
| | - Jing Liu
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 ShuaiFuYuan Street, Dongcheng District, Beijing, 100730, China
| | - Juan Xiao
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 ShuaiFuYuan Street, Dongcheng District, Beijing, 100730, China.
| | - Hongmei Song
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 ShuaiFuYuan Street, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
10
|
Nombel A, Fabien N, Coutant F. Dermatomyositis With Anti-MDA5 Antibodies: Bioclinical Features, Pathogenesis and Emerging Therapies. Front Immunol 2021; 12:773352. [PMID: 34745149 PMCID: PMC8564476 DOI: 10.3389/fimmu.2021.773352] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/07/2021] [Indexed: 12/24/2022] Open
Abstract
Anti-MDA5 dermatomyositis is a rare systemic autoimmune disease, historically described in Japanese patients with clinically amyopathic dermatomyositis and life-threatening rapidly progressive interstitial lung disease. Subsequently, the complete clinical spectrum of the disease was enriched by skin, articular and vascular manifestations. Depending on the predominance of these symptoms, three distinct clinical phenotypes with different prognosis are now defined. To date, the only known molecular component shared by the three entities are specific antibodies targeting MDA5, a cytosolic protein essential for antiviral host immune responses. Several biological tools have emerged to detect these antibodies, with drawbacks and limitations for each of them. However, the identification of this highly specific serological marker of the disease raises the question of its role in the pathogenesis. Although current knowledge on the pathogenic mechanisms that take place in the disease are still in their enfancy, several lines of evidence support a central role of interferon-mediated vasculopathy in the development of skin and lung lesions, as well as a possible pathogenic involvement of anti-MDA5 antibodies. Here, we review the clinical and biological evidences in favor of these hypothesis, and we discuss the contribution of emerging therapies that shed some light on the pathogenesis of the disease.
Collapse
Affiliation(s)
- Anaïs Nombel
- Immunology Department, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Nicole Fabien
- Immunology Department, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Frédéric Coutant
- Immunology Department, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre-Bénite, France.,Immunogenomics and Inflammation Research Team, University of Lyon, Edouard Herriot Hospital, Lyon, France
| |
Collapse
|
11
|
Szumilas N, Corneth OBJ, Lehmann CHK, Schmitt H, Cunz S, Cullen JG, Chu T, Marosan A, Mócsai A, Benes V, Zehn D, Dudziak D, Hendriks RW, Nitschke L. Siglec-H-Deficient Mice Show Enhanced Type I IFN Responses, but Do Not Develop Autoimmunity After Influenza or LCMV Infections. Front Immunol 2021; 12:698420. [PMID: 34497606 PMCID: PMC8419311 DOI: 10.3389/fimmu.2021.698420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/27/2021] [Indexed: 12/02/2022] Open
Abstract
Siglec-H is a DAP12-associated receptor on plasmacytoid dendritic cells (pDCs) and microglia. Siglec-H inhibits TLR9-induced IFN-α production by pDCs. Previously, it was found that Siglec-H-deficient mice develop a lupus-like severe autoimmune disease after persistent murine cytomegalovirus (mCMV) infection. This was due to enhanced type I interferon responses, including IFN-α. Here we examined, whether other virus infections can also induce autoimmunity in Siglec-H-deficient mice. To this end we infected Siglec-H-deficient mice with influenza virus or with Lymphocytic Choriomeningitis virus (LCMV) clone 13. With both types of viruses we did not observe induction of autoimmune disease in Siglec-H-deficient mice. This can be explained by the fact that both types of viruses are ssRNA viruses that engage TLR7, rather than TLR9. Also, Influenza causes an acute infection that is rapidly cleared and the chronicity of LCMV clone 13 may not be sufficient and may rather suppress pDC functions. Siglec-H inhibited exclusively TLR-9 driven type I interferon responses, but did not affect type II or type III interferon production by pDCs. Siglec-H-deficient pDCs showed impaired Hck expression, which is a Src-family kinase expressed in myeloid cells, and downmodulation of the chemokine receptor CCR9, that has important functions for pDCs. Accordingly, Siglec-H-deficient pDCs showed impaired migration towards the CCR9 ligand CCL25. Furthermore, autoimmune-related genes such as Klk1 and DNase1l3 are downregulated in Siglec-H-deficient pDCs as well. From these findings we conclude that Siglec-H controls TLR-9-dependent, but not TLR-7 dependent inflammatory responses after virus infections and regulates chemokine responsiveness of pDCs.
Collapse
Affiliation(s)
- Nadine Szumilas
- Division of Genetics, Department of Biology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Odilia B J Corneth
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Christian H K Lehmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Erlangen, Germany.,Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, University of Erlangen-Nürnberg, Erlangen, Germany.,Medical Immunology Campus Erlangen (MICE), University of Erlangen-Nürnberg, Erlangen, Germany
| | - Heike Schmitt
- First Department of Medicine, University Hospital Erlangen, Erlangen, Germany
| | - Svenia Cunz
- Division of Genetics, Department of Biology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Jolie G Cullen
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Talyn Chu
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Anita Marosan
- Department of Immune Modulation, University Hospital Erlangen, Erlangen, Germany
| | - Attila Mócsai
- Semmelweis University School of Medicine, Budapest, Hungary
| | - Vladimir Benes
- Genomics Core Facility, EMBL Heidelberg, Heidelberg, Germany
| | - Dietmar Zehn
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Erlangen, Germany.,Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, University of Erlangen-Nürnberg, Erlangen, Germany.,Medical Immunology Campus Erlangen (MICE), University of Erlangen-Nürnberg, Erlangen, Germany
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Lars Nitschke
- Division of Genetics, Department of Biology, University of Erlangen-Nürnberg, Erlangen, Germany.,Medical Immunology Campus Erlangen (MICE), University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
12
|
Tarantino G, Citro V, Cataldi M. Findings from Studies Are Congruent with Obesity Having a Viral Origin, but What about Obesity-Related NAFLD? Viruses 2021; 13:1285. [PMID: 34372491 PMCID: PMC8310150 DOI: 10.3390/v13071285] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/26/2021] [Accepted: 06/26/2021] [Indexed: 02/06/2023] Open
Abstract
Infection has recently started receiving greater attention as an unusual causative/inducing factor of obesity. Indeed, the biological plausibility of infectobesity includes direct roles of some viruses to reprogram host metabolism toward a more lipogenic and adipogenic status. Furthermore, the probability that humans may exchange microbiota components (virome/virobiota) points out that the altered response of IFN and other cytokines, which surfaces as a central mechanism for adipogenesis and obesity-associated immune suppression, is due to the fact that gut microbiota uphold intrinsic IFN signaling. Last but not least, the adaptation of both host immune and metabolic system under persistent viral infections play a central role in these phenomena. We hereby discuss the possible link between adenovirus and obesity-related nonalcoholic fatty liver disease (NAFLD). The mechanisms of adenovirus-36 (Ad-36) involvement in hepatic steatosis/NAFLD consist in reducing leptin gene expression and insulin sensitivity, augmenting glucose uptake, activating the lipogenic and pro-inflammatory pathways in adipose tissue, and increasing the level of macrophage chemoattractant protein-1, all of these ultimately leading to chronic inflammation and altered lipid metabolism. Moreover, by reducing leptin expression and secretion Ad-36 may have in turn an obesogenic effect through increased food intake or decreased energy expenditure via altered fat metabolism. Finally, Ad-36 is involved in upregulation of cAMP, phosphatidylinositol 3-kinase, and p38 signaling pathways, downregulation of Wnt10b expression, increased expression of CCAAT/enhancer binding protein-beta, and peroxisome proliferator-activated receptor gamma 2 with consequential lipid accumulation.
Collapse
Affiliation(s)
- Giovanni Tarantino
- Department of Clinical Medicine and Surgery, “Federico II” University Medical School of Naples, 80131 Napoli, Italy
| | - Vincenzo Citro
- Department of General Medicine, “Umberto I” Hospital, Nocera Inferiore (Sa), 84014 Nocera Inferiore, Italy;
| | - Mauro Cataldi
- Section of Pharmacology, Department of Neuroscience, Reproductive Sciences and Dentistry, “Federico II” University of Naples, 80131 Napoli, Italy;
| |
Collapse
|
13
|
Manunta MDI, Lamorte G, Ferrari F, Trombetta E, Tirone M, Bianco C, Cattaneo A, Santoro L, Baselli G, Brasca M, Ostadreza M, Erba E, Gori A, Bandera A, Porretti L, Valenti LVC, Prati D. Impact of SARS-CoV-2 infection on the recovery of peripheral blood mononuclear cells by density gradient. Sci Rep 2021; 11:4904. [PMID: 33649400 PMCID: PMC7921094 DOI: 10.1038/s41598-021-83950-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
SARS-CoV-2 virus infection is responsible for coronavirus disease (COVID-19), which is characterised by a hyperinflammatory response that plays a major role in determining the respiratory and immune-mediated complications of this condition. While isolating peripheral blood mononuclear cells (PBMCs) from whole blood of COVID-19 patients by density gradient centrifugation, we noticed some changes in the floating properties and in the sedimentation of the cells on density medium. Investigating this further, we found that in early phase COVID-19 patients, characterised by reduced circulating lymphocytes and monocytes, the PBMC fraction contained surprisingly high levels of neutrophils. Furthermore, the neutrophil population exhibited alterations in the cell size and in the internal complexity, consistent with the presence of low density neutrophils (LDNs) and immature forms, which may explain the shift seen in the floating abilities and that may be predictive of the severity of the disease. The percentage of this subset of neutrophils found in the PBMC band was rather spread (35.4 ± 27.2%, with a median 28.8% and IQR 11.6-56.1, Welch's t-test early phase COVID-19 versus blood donor healthy controls P < 0.0001). Results confirm the presence of an increased number of LDNs in patients with early stage COVID-19, which correlates with disease severity and may be recovered by centrifugation on a density gradient together with PBMCs.
Collapse
Affiliation(s)
- Maria D I Manunta
- Department of Transfusion Medicine and Hematology, Milano Cord Blood Bank, Processing Facility and Biobank POLI-MI, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza, 35, 20122, Milan, Italy.
| | - Giuseppe Lamorte
- Department of Transfusion Medicine and Hematology, Milano Cord Blood Bank, Processing Facility and Biobank POLI-MI, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza, 35, 20122, Milan, Italy
| | - Francesca Ferrari
- Department of Transfusion Medicine and Hematology, Milano Cord Blood Bank, Processing Facility and Biobank POLI-MI, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza, 35, 20122, Milan, Italy
| | - Elena Trombetta
- Flow Cytometry and Cell Sorting Laboratory, Analysis Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Mario Tirone
- Flow Cytometry and Cell Sorting Laboratory, Analysis Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Cristiana Bianco
- Department of Transfusion Medicine and Hematology, Milano Cord Blood Bank, Processing Facility and Biobank POLI-MI, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza, 35, 20122, Milan, Italy
| | - Alessandra Cattaneo
- Flow Cytometry and Cell Sorting Laboratory, Analysis Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luigi Santoro
- Department of Transfusion Medicine and Hematology, Milano Cord Blood Bank, Processing Facility and Biobank POLI-MI, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza, 35, 20122, Milan, Italy
| | - Guido Baselli
- Department of Transfusion Medicine and Hematology, Milano Cord Blood Bank, Processing Facility and Biobank POLI-MI, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza, 35, 20122, Milan, Italy
| | - Manuela Brasca
- Department of Transfusion Medicine and Hematology, Milano Cord Blood Bank, Processing Facility and Biobank POLI-MI, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza, 35, 20122, Milan, Italy.,Directorate of Allied Health Professions, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Mahnoosh Ostadreza
- Department of Transfusion Medicine and Hematology, Milano Cord Blood Bank, Processing Facility and Biobank POLI-MI, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza, 35, 20122, Milan, Italy
| | - Elisa Erba
- Department of Transfusion Medicine and Hematology, Milano Cord Blood Bank, Processing Facility and Biobank POLI-MI, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza, 35, 20122, Milan, Italy.,Directorate of Allied Health Professions, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Andrea Gori
- Infectious Diseases Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Centre for Multidisciplinary Research in Health Science (MACH), Università degli Studi di Milano, Milan, Italy
| | - Alessandra Bandera
- Infectious Diseases Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Centre for Multidisciplinary Research in Health Science (MACH), Università degli Studi di Milano, Milan, Italy
| | - Laura Porretti
- Flow Cytometry and Cell Sorting Laboratory, Analysis Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca V C Valenti
- Department of Transfusion Medicine and Hematology, Milano Cord Blood Bank, Processing Facility and Biobank POLI-MI, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza, 35, 20122, Milan, Italy.,Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Daniele Prati
- Department of Transfusion Medicine and Hematology, Milano Cord Blood Bank, Processing Facility and Biobank POLI-MI, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza, 35, 20122, Milan, Italy
| |
Collapse
|
14
|
Georgiev T, Angelov AK. Complexities of diagnosis and management of COVID-19 in autoimmune diseases: Potential benefits and detriments of immunosuppression. World J Clin Cases 2020; 8:3669-3678. [PMID: 32953843 PMCID: PMC7479565 DOI: 10.12998/wjcc.v8.i17.3669] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/12/2020] [Accepted: 08/26/2020] [Indexed: 02/05/2023] Open
Abstract
Recent advances in our understanding of coronavirus disease 2019 (COVID-19) and the associated acute respiratory distress syndrome might approximate the cytokine release syndrome of severe immune-mediated disease. Importantly, this presumption provides the rationale for utilization of therapy, until recently reserved mostly for autoimmune diseases (ADs), in the management of COVID-19 hyperinflammation condition and has led to an extensive discussion for the potential benefits and detriments of immunosuppression. Our paper intends to examine the available recommendations, complexities in diagnosis and management when dealing with patients with ADs amidst the COVID-19 crisis. Mimicking a flare of an underlying AD, overlapping pathological lung patterns, probability of higher rates of false-positive antibody test, and lack of concrete data are only a part of the detrimental and specific characteristics of COVID-19 outbreak among the population with ADs. The administration of pharmaceutical therapy should not undermine the physical and psychological status of the patient with the maximum utilization of telemedicine. Researchers and clinicians should be vigilant for upcoming research for insight and perspective to fine-tune the clinical guidelines and practice and to weigh the potential benefits and detrimental effects of the applied immunomodulating therapy.
Collapse
Affiliation(s)
- Tsvetoslav Georgiev
- Clinic of Rheumatology, University Hospital "St. Marina", First Department of Internal Medicine, Medical University - Varna, Varna 9010, Bulgaria
| | | |
Collapse
|
15
|
Zhu FX, Huang JY, Ye Z, Wen QQ, Wei JCC. Risk of systemic lupus erythematosus in patients with idiopathic thrombocytopenic purpura: a population-based cohort study. Ann Rheum Dis 2020; 79:793-799. [PMID: 32241798 DOI: 10.1136/annrheumdis-2020-217013] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 11/04/2022]
Abstract
BACKGROUND Idiopathic thrombocytopenic purpura (ITP) may play a role in early-stage systemic lupus erythematosus (SLE). The incidence of SLE in patients with ITP and the potential relationship between them is still unclear. This study was performed to provide epidemiological evidence regarding the relationship between ITP and SLE occurrence. METHODS In this population-based retrospective cohort study, the risk of SLE was analysed in a cohort of patients newly diagnosed with ITP between 2000 and 2013. Controls were selected at a 1:2 ratio through propensity score matching (PSM) using the greedy algorithm. The Cox proportional hazard model was used to analyse the association between ITP and SLE incidence. There were four different Cox regression models, and the sensitivity analyses were implemented to evaluate the HR of SLE after exposure with ITP. RESULTS In the age-matched and sex-matched ITP and non-ITP cohort, the average follow-up time was about 80 months in this study. There were 34 (4.70%) and 27 (0.19%) incident cases of SLE in ITP and non-ITP group. The incidence rates were 62.0 (95% CI 44.3 to 86.8) and 2.10 (95% CI 1.44 to 3.06), respectively. The adjusted HR of incidental SLE in the ITP group was 25.1 (95% CI 13.7 to 46.0). The other risk factors for SLE were female sex and Sjogren's syndrome. After PSM, the incidence rate and Kaplan-Meir curves of SLE were consistent with the results for the age-matched and sex-matched population, the HR 17.4 (95% CI 5.28 to 57.4) was estimated by conditional Cox model. CONCLUSION This cohort study demonstrated that patients with ITP have a higher risk of SLE. Clinically, patients with ITP should be monitored for incidental lupus.
Collapse
Affiliation(s)
- Fang-Xiao Zhu
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Jing-Yang Huang
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung City, Taiwan, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Zhizhong Ye
- Department of Rheumatology, Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, Guangdong, China
| | - Qing-Qing Wen
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - James Cheng-Chung Wei
- Department of Rheumatology, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Institute of Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Chung Shan Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
16
|
Björk A, Mofors J, Wahren-Herlenius M. Environmental factors in the pathogenesis of primary Sjögren's syndrome. J Intern Med 2020; 287:475-492. [PMID: 32107824 DOI: 10.1111/joim.13032] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Primary Sjögren's syndrome (SS) is a systemic autoimmune disease in which exocrine organs, primarily the salivary and lacrimal glands, are targets of chronic inflammation, leading to severe dryness of eyes and mouth. Fatigue and arthralgia are also common, and extraglandular manifestations involving the respiratory, nervous and vascular systems occur in a subset of patients. Persistent activation of the type I interferon system, and autoreactive B and T cells with production of disease-associated autoantibodies are central to the pathogenesis. Genetic polymorphisms that associate with an increased risk of SS have been described, though the risk-increase contributed by the respective variant is generally low. It is thus becoming increasingly clear that genetics cannot alone account for the development of SS and that other, presumably exogenous, factors must play a critical role. Relatively few studies have investigated exposure to potential risk factors prior to SS disease onset. Rather, many factors have been studied in prevalent cases. In this review, we summarize current literature on exogenous factors in the pathogenesis of SS including infections, hormones, smoking, solvents and additional compounds. We delineate for which factors there is current evidence of increased disease risk, and for which our present knowledge is confined to suggesting their role in SS pathogenesis. Finally, we outline future perspectives in the continued search for environmental risk factors for SS, a research area of great importance considering the possibilities for preventive measures.
Collapse
Affiliation(s)
- A Björk
- From the, Division of Rheumatology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - J Mofors
- From the, Division of Rheumatology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - M Wahren-Herlenius
- From the, Division of Rheumatology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
17
|
Espéli M, Bashford-Rogers R, Sowerby JM, Alouche N, Wong L, Denton AE, Linterman MA, Smith KGC. FcγRIIb differentially regulates pre-immune and germinal center B cell tolerance in mouse and human. Nat Commun 2019; 10:1970. [PMID: 31036800 PMCID: PMC6488660 DOI: 10.1038/s41467-019-09434-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 02/21/2019] [Indexed: 11/18/2022] Open
Abstract
Several tolerance checkpoints exist throughout B cell development to control autoreactive B cells and prevent the generation of pathogenic autoantibodies. FcγRIIb is an Fc receptor that inhibits B cell activation and, if defective, is associated with autoimmune disease, yet its impact on specific B cell tolerance checkpoints is unknown. Here we show that reduced expression of FcγRIIb enhances the deletion and anergy of autoreactive immature B cells, but in contrast promotes autoreactive B cell expansion in the germinal center and serum autoantibody production, even in response to exogenous, non-self antigens. Our data thus show that FcγRIIb has opposing effects on pre-immune and post-immune tolerance checkpoints, and suggest that B cell tolerance requires the control of bystander germinal center B cells with low or no affinity for the immunizing antigen. The inhibitory receptor, FcγRIIb, is reported to limit autoimmune B cell response. Here the authors show that FcγRIIb has a dual role in both human and mouse, with reduced FcγRIIb expression or function associated with enhanced pre-immune B cell tolerance, yet defective control of mature autoreactive B cells in the germinal center.
Collapse
Affiliation(s)
- Marion Espéli
- The Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 OXY, England, UK. .,UMR996 - Inflammation, Chemokines and Immunopathology, Inserm, Univ Paris-Sud, Université Paris-Saclay, Clamart, F-92140, France.
| | - Rachael Bashford-Rogers
- The Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 OXY, England, UK.,Wellcome Centre for Human Genetics, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - John M Sowerby
- The Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 OXY, England, UK.,Cambridge Institute of Therapeutic Immunology & Infectious Disease, Jeffrey Cheah Biomedical Centre Cambridge Biomedical Campus, University of Cambridge, CB2 0AW, Cambridge, UK
| | - Nagham Alouche
- UMR996 - Inflammation, Chemokines and Immunopathology, Inserm, Univ Paris-Sud, Université Paris-Saclay, Clamart, F-92140, France
| | - Limy Wong
- The Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 OXY, England, UK
| | - Alice E Denton
- The Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 OXY, England, UK.,Lymphocyte Signalling and Development, Babraham Institute, CB22 3AT, Cambridge, UK
| | - Michelle A Linterman
- The Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 OXY, England, UK.,Lymphocyte Signalling and Development, Babraham Institute, CB22 3AT, Cambridge, UK
| | - Kenneth G C Smith
- The Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 OXY, England, UK. .,Cambridge Institute of Therapeutic Immunology & Infectious Disease, Jeffrey Cheah Biomedical Centre Cambridge Biomedical Campus, University of Cambridge, CB2 0AW, Cambridge, UK.
| |
Collapse
|
18
|
Dinse GE, Parks CG, Meier HCS, Co CA, Chan EKL, Jusko TA, Yeh J, Miller FW. Prescription medication use and antinuclear antibodies in the United States, 1999-2004. J Autoimmun 2018; 92:93-103. [PMID: 29779929 PMCID: PMC6054905 DOI: 10.1016/j.jaut.2018.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/02/2018] [Accepted: 05/10/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND Clinical reports link specific medications with the development of antinuclear antibodies (ANA), but population-based evidence is limited. OBJECTIVE The present study investigated associations between prescription medication use and ANA in a representative sample of the adult noninstitutionalized US population, first focusing on medications previously related to ANA and then considering all medications reported in the National Health and Nutrition Examination Survey (NHANES). METHODS Based on NHANES data (1999-2004) for 3608 adults (ages ≥18 years), we estimated odds ratios (ORs) and 95% confidence intervals (CIs) to assess associations between recent medication use and ANA (overall and in sex and age subgroups), adjusted for potential confounders and the survey sampling design. RESULTS We found no evidence that most medications previously associated with ANA in specific individuals were risk factors for ANA in the general population, although statistical power was limited for some medications. Overall, ANA were less prevalent in adults who recently used any prescription medications compared with those who did not (OR = 0.73; CI = 0.57,0.93), and likewise several classes of medications were inversely associated with ANA, including hormones (OR = 0.73; CI = 0.55,0.98), thiazide diuretics (OR = 0.43; CI = 0.24,0.79), sulfonylureas (OR = 0.41; CI = 0.19,0.89), and selective serotonin reuptake inhibitor antidepressants (OR = 0.65; CI = 0.42,0.98). Positive associations with ANA were seen for loop diuretics (OR = 1.72; CI = 1.03,2.88) in all adults, and for benzodiazepines (OR = 2.11; CI = 1.09,4.10) and bronchodilators (OR = 1.83; CI = 1.00,3.38) in older (ages ≥60) adults. Estrogens were positively associated with ANA in older women (OR = 1.80; CI = 1.00,3.23) but inversely associated with ANA in younger (ages 18-59) women (OR = 0.43; CI = 0.20,0.93). Regarding individual medications, ANA were positively associated with ciprofloxacin (OR = 4.23; CI = 1.21,14.8), furosemide (OR = 1.79; CI = 1.09,2.93), and omeprazole (OR = 2.05; CI = 1.03,4.10) in all adults, and with salmeterol (OR = 3.76; CI = 1.66,8.52), tolterodine (OR = 6.64; CI = 1.45,30.5), and triamterene (OR = 3.10; CI = 1.08,8.88) in older adults. Also, in younger adults, hydrochlorothiazide was inversely associated with ANA (OR = 0.44; CI = 0.20,0.98). CONCLUSIONS Our findings in the general population do not confirm most clinically reported positive associations between specific medications and ANA in some individuals. However, novel positive ANA associations with other medications, as well as unexplained inverse associations with certain classes of medications and overall medication use, deserve further research to clarify the possible roles of medications as risk and protective factors in the development of autoantibodies and autoimmune disease.
Collapse
Affiliation(s)
- Gregg E Dinse
- Public Health Sciences, Social & Scientific Systems Inc., Durham, NC, USA.
| | - Christine G Parks
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA.
| | - Helen C S Meier
- Joseph J. Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI, USA.
| | - Caroll A Co
- Public Health Sciences, Social & Scientific Systems Inc., Durham, NC, USA.
| | - Edward K L Chan
- University of Florida Health Science Center, Gainesville, FL, USA.
| | - Todd A Jusko
- Departments of Public Health Sciences and Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| | - James Yeh
- Department of Anesthesiology, Perioperative and Pain Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA,.
| | - Frederick W Miller
- Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA.
| |
Collapse
|
19
|
|
20
|
Netchiporouk E, Sasseville D, Moreau L, Habel Y, Rahme E, Ben-Shoshan M. Evaluating Comorbidities, Natural History, and Predictors of Early Resolution in a Cohort of Children With Chronic Urticaria. JAMA Dermatol 2017; 153:1236-1242. [PMID: 28973060 DOI: 10.1001/jamadermatol.2017.3182] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance Chronic urticaria (CU) affects 0.1% to 0.3% of children. Most cases have no identifiable trigger and are classified as chronic spontaneous urticaria (CSU). At least half of patients with CSU may have an autoimmune etiology that can be determined in vitro using the basophil activation test (BAT). While 30% to 55% of CU cases resolve spontaneously within 5 years in adults, the natural history and predictors of resolution in children are not known. Objective To assess the comorbidities, natural history of CU, and its subtypes in children and identify predictors of resolution. Design, Setting, and Participants We followed a pediatric cohort with chronic urticaria that presented with hives lasting at least 6 weeks between 2013 and 2015 at a single tertiary care referral center. Exposures Data were collected on disease activity, comorbidities, physical triggers, BAT results, complete blood cell count, C-reactive protein levels, thyroid-stimulating hormone levels, and thyroid peroxidase antibodies. Main Outcomes and Measures We assessed the rate of resolution (defined as absence of hives for at least 1 year with no treatment) and the association with clinical and laboratory markers. Results The cohort comprised 139 children younger than 18 years old. Thirty-one patients (20%) had inducible urticaria, most commonly cold induced. Six children had autoimmune comorbidity, such as thyroiditis and type 1 diabetes. Autoimmune disorders (24 patients [17%]) and CU (17 patients [12%]) were common in family members. Positive BAT results (CD63 levels > 1.8%) were found in 58% of patients. Patients with positive BAT results (CD63 level >1.8%) were twice as likely to resolve after 1 year compared with negative BAT results (hazard ratio [HR], 2.33; 95% CI, 1.08-5.05). In contrast, presence of basophils decreased the likelihood of resolution (HR, 0.40; 95% CI, 0.20-0.99). No correlation with age was found. Chronic urticaria resolved in 43 patients, with a rate of resolution of 10.3% per year. Levels of CD63 higher than 1.8% and absence of basophils were associated with earlier disease resolution. Conclusions and Relevance Resolution rate in children with CU is low. The presence of certain biomarkers (positive BAT result and basophil count) may help to predict the likelihood of resolution.
Collapse
Affiliation(s)
- Elena Netchiporouk
- Division of Dermatology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Denis Sasseville
- Division of Dermatology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Linda Moreau
- Division of Dermatology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Youssef Habel
- Division of Clinical Epidemiology, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| | - Elham Rahme
- Division of Clinical Epidemiology, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| | - Moshe Ben-Shoshan
- Division of Allergy and Clinical Immunology, Department of Pediatrics, McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
21
|
Abstract
Autoantibody testing is performed to help diagnose patients who have clinical symptoms suggestive of possible autoimmune diseases. Antinuclear antibodies (ANA) are present in many systemic autoimmune conditions such as systemic lupus erythematosus (SLE). However, a positive ANA test may also be seen with non-autoimmune inflammatory diseases, including both acute and chronic infections. When the ANA test is used as an initial screen in patients with non-specific clinical symptoms, such as fever, joint pain, myalgias, fatigue, rash, or anemia, the likelihood of a positive result due to infection will increase, especially in children. This article identifies acute and chronic infectious diseases that are likely to produce a positive ANA result and summarizes recent literature addressing both the causes and consequences of these findings.
Collapse
Affiliation(s)
- Christine M Litwin
- a Pathology and Laboratory Medicine, Medical University of South Carolina , Charleston , South Carolina , USA
| | - Steven R Binder
- b Clinical Diagnostics Group, Bio-Rad Laboratories , Hercules , California , USA
| |
Collapse
|
22
|
Abstract
Parvovirus B19 (B19V) and human bocavirus 1 (HBoV1), members of the large Parvoviridae family, are human pathogens responsible for a variety of diseases. For B19V in particular, host features determine disease manifestations. These viruses are prevalent worldwide and are culturable in vitro, and serological and molecular assays are available but require careful interpretation of results. Additional human parvoviruses, including HBoV2 to -4, human parvovirus 4 (PARV4), and human bufavirus (BuV) are also reviewed. The full spectrum of parvovirus disease in humans has yet to be established. Candidate recombinant B19V vaccines have been developed but may not be commercially feasible. We review relevant features of the molecular and cellular biology of these viruses, and the human immune response that they elicit, which have allowed a deep understanding of pathophysiology.
Collapse
Affiliation(s)
- Jianming Qiu
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | | | - Neal S Young
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
23
|
Abdel-Wahab N, Lopez-Olivo MA, Pinto-Patarroyo GP, Suarez-Almazor ME. Systematic review of case reports of antiphospholipid syndrome following infection. Lupus 2016; 25:1520-1531. [PMID: 27060064 PMCID: PMC7508159 DOI: 10.1177/0961203316640912] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 02/29/2016] [Indexed: 11/17/2022]
Abstract
OBJECTIVE The objective of this study was to conduct a systematic review of case reports documenting the development of antiphospholipid syndrome or antiphospholipid syndrome-related features after an infection. METHODS We searched Medline, EMBASE, Web of Science, PubMed ePubs, and The Cochrane Library - CENTRAL through March 2015 without restrictions. Studies reporting cases of antiphospholipid syndrome or antiphospholipid syndrome-related features following an infection were included. RESULTS Two hundred and fifty-nine publications met inclusion criteria, reporting on 293 cases. Three different groups of patients were identified; group 1 included patients who fulfilled the criteria for definitive antiphospholipid syndrome (24.6%), group 2 included patients who developed transient antiphospholipid antibodies with thromboembolic phenomena (43.7%), and group 3 included patients who developed transient antiphospholipid antibodies without thromboembolic events (31.7%). The most common preceding infection was viral (55.6%). In cases that developed thromboembolic events Human immunodeficiency and Hepatitis C viruses were the most frequently reported. Parvovirus B19 was the most common in cases that developed antibodies without thromboembolic events. Hematological manifestations and peripheral thrombosis were the most common clinical manifestations. Positive anticardiolipin antibodies were the most frequent antibodies reported, primarily coexisting IgG and IgM isotypes. Few patients in groups 1 and 2 had persistent antiphospholipid antibodies for more than 6 months. Outcome was variable with some cases reporting persistent antiphospholipid syndrome features and others achieving complete resolution of clinical events. CONCLUSIONS Development of antiphospholipid antibodies with all traditional manifestations of antiphospholipid syndrome were observed after variety of infections, most frequently after chronic viral infections with Human immunodeficiency and Hepatitis C. The causal relationship between infection and antiphospholipid syndrome cannot be established, but the possible contribution of various infections in the pathogenesis of antiphospholipid syndrome need further longitudinal and controlled studies to establish the incidence, and better quantify the risk and the outcomes of antiphospholipid-related events after infection.
Collapse
Affiliation(s)
- N Abdel-Wahab
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Rheumatology and Rehabilitation Department, Assiut University Hospitals, Assiut, Egypt
| | - M A Lopez-Olivo
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - G P Pinto-Patarroyo
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - M E Suarez-Almazor
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
24
|
Schönrich G, Raftery MJ. Neutrophil Extracellular Traps Go Viral. Front Immunol 2016; 7:366. [PMID: 27698656 PMCID: PMC5027205 DOI: 10.3389/fimmu.2016.00366] [Citation(s) in RCA: 191] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 09/05/2016] [Indexed: 12/14/2022] Open
Abstract
Neutrophils are the most numerous immune cells. Their importance as the first line of defense against bacterial and fungal pathogens is well described. In contrast, the role of neutrophils in controlling viral infections is less clear. Bacterial and fungal pathogens can stimulate neutrophils extracellular traps (NETs) in a process called NETosis. Although NETosis has previously been described as a special form of programmed cell death, there are forms of NET production that do not end with the demise of neutrophils. As an end result of NETosis, genomic DNA complexed with microbicidal proteins is expelled from neutrophils. These structures can kill pathogens or at least prevent their local spread within host tissue. On the other hand, disproportionate NET formation can cause local or systemic damage. Only recently, it was recognized that viruses can also induce NETosis. In this review, we discuss the mechanisms by which NETs are produced in the context of viral infection and how this may contribute to both antiviral immunity and immunopathology. Finally, we shed light on viral immune evasion mechanisms targeting NETs.
Collapse
Affiliation(s)
- Günther Schönrich
- Institute of Medical Virology, Helmut-Ruska-Haus, Charité - Universitätsmedizin Berlin , Berlin , Germany
| | - Martin J Raftery
- Institute of Medical Virology, Helmut-Ruska-Haus, Charité - Universitätsmedizin Berlin , Berlin , Germany
| |
Collapse
|
25
|
Schmitt H, Sell S, Koch J, Seefried M, Sonnewald S, Daniel C, Winkler TH, Nitschke L. Siglec-H protects from virus-triggered severe systemic autoimmunity. J Exp Med 2016; 213:1627-44. [PMID: 27377589 PMCID: PMC4986536 DOI: 10.1084/jem.20160189] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 05/13/2016] [Indexed: 12/23/2022] Open
Abstract
Siglec-H is a key negative regulator of the type I interferon pathway, reducing the incidence of autoimmunity after viral infection. It is controversial whether virus infections can contribute to the development of autoimmune diseases. Type I interferons (IFNs) are critical antiviral cytokines during virus infections and have also been implicated in the pathogenesis of systemic lupus erythematosus. Type I IFN is mainly produced by plasmacytoid dendritic cells (pDCs). The secretion of type I IFN of pDCs is modulated by Siglec-H, a DAP12-associated receptor on pDCs. In this study, we show that Siglec-H–deficient pDCs produce more of the type I IFN, IFN-α, in vitro and that Siglec-H knockout (KO) mice produce more IFN-α after murine cytomegalovirus (mCMV) infection in vivo. This did not impact control of viral replication. Remarkably, several weeks after a single mCMV infection, Siglec-H KO mice developed a severe form of systemic lupus–like autoimmune disease with strong kidney nephritis. In contrast, uninfected aging Siglec-H KO mice developed a mild form of systemic autoimmunity. The induction of systemic autoimmune disease after virus infection in Siglec-H KO mice was accompanied by a type I IFN signature and fully dependent on type I IFN signaling. These results show that Siglec-H normally serves as a modulator of type I IFN responses after infection with a persistent virus and thereby prevents induction of autoimmune disease.
Collapse
Affiliation(s)
- Heike Schmitt
- Division of Genetics, Department of Biology, University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Sabrina Sell
- Nikolaus-Fiebiger-Zentrum, Division of Genetics, Department of Biology, University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Julia Koch
- Division of Genetics, Department of Biology, University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Martina Seefried
- Nikolaus-Fiebiger-Zentrum, Division of Genetics, Department of Biology, University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Sophia Sonnewald
- Division of Biochemistry, Department of Biology, University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Christoph Daniel
- Department of Nephropathology, University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Thomas H Winkler
- Nikolaus-Fiebiger-Zentrum, Division of Genetics, Department of Biology, University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Lars Nitschke
- Division of Genetics, Department of Biology, University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
26
|
The quest for autoreactive antibodies in nasal polyps. J Allergy Clin Immunol 2016; 138:893-895.e5. [PMID: 27283383 DOI: 10.1016/j.jaci.2016.03.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 03/04/2016] [Accepted: 03/21/2016] [Indexed: 11/21/2022]
|
27
|
Serology of Lupus Erythematosus: Correlation between Immunopathological Features and Clinical Aspects. Autoimmune Dis 2014; 2014:321359. [PMID: 24649358 PMCID: PMC3932647 DOI: 10.1155/2014/321359] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 12/23/2013] [Indexed: 02/06/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the aberrant production of a broad and heterogenous group of autoantibodies. Even though the presence of autoantibodies in SLE has been known, for more than 60 years, still nowadays a great effort is being made to understand the pathogenetic, diagnostic, and prognostic meaning of such autoantibodies.
Antibodies to ds-DNA are useful for the diagnosis of SLE, to monitor the disease activity, and correlate with renal and central nervous involvements. Anti-Sm antibodies are highly specific for SLE. Anti-nucleosome antibodies are an excellent marker for SLE and good predictors of flares in quiescent lupus. Anti-histone antibodies characterize drug-induced lupus, while anti-SSA/Ro and anti-SSB/La antibodies are associated with neonatal lupus erythematosus and photosensitivity. Anti-ribosomal P antibodies play a role in neuropsychiatric lupus, but their association with clinical manifestations is still unclear. Anti-phospholipid antibodies are associated with the anti-phospholipid syndrome, cerebral vascular disease, and neuropsychiatric lupus. Anti-C1q antibodies amplify glomerular injury, and the elevation of their titers may predict renal flares. Anti-RNP antibodies are a marker of Sharp's syndrome but can be found in SLE as well. Anti-PCNA antibodies are present in 5–10% of SLE patients especially those with arthritis and hypocomplementemia.
Collapse
|
28
|
Hayashi N, Koshiba M, Nishimura K, Sugiyama D, Nakamura T, Morinobu S, Kawano S, Kumagai S. Prevalence of disease-specific antinuclear antibodies in general population: estimates from annual physical examinations of residents of a small town over a 5-year period. Mod Rheumatol 2014. [DOI: 10.3109/s10165-008-0028-1] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
29
|
Treatment of systemic lupus erythematosus: new therapeutic avenues and blind alleys. Nat Rev Rheumatol 2013; 10:23-34. [DOI: 10.1038/nrrheum.2013.145] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
30
|
Fox A, Hung TM, Wertheim H, Hoa LNM, Vincent A, Lang B, Waters P, Ha NH, Trung NV, Farrar J, Van Kinh N, Horby P. Acute measles encephalitis in partially vaccinated adults. PLoS One 2013; 8:e71671. [PMID: 23967232 PMCID: PMC3742472 DOI: 10.1371/journal.pone.0071671] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 07/08/2013] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND The pathogenesis of acute measles encephalitis (AME) is poorly understood. Treatment with immune-modulators is based on theories that post-infectious autoimmune responses cause demyelination. The clinical course and immunological parameters of AME were examined during an outbreak in Vietnam. METHODS AND FINDINGS Fifteen measles IgM-positive patients with confusion or Glasgow Coma Scale (GCS) score below 13, and thirteen with uncomplicated measles were enrolled from 2008-2010. Standardized clinical exams were performed and blood collected for lymphocyte and measles- and auto-antibody analysis. The median age of AME patients was 21 years, similar to controls. Eleven reported receiving measles vaccination when aged one year. Confusion developed a median of 4 days after rash. Six patients had GCS <8 and four required mechanical ventilation. CSF showed pleocytosis (64%) and proteinorrhachia (71%) but measles virus RNA was not detected. MRI revealed bilateral lesions in the cerebellum and brain stem in some patients. Most received dexamethasone +/- IVIG within 4 days of admission but symptoms persisted for ≥3 weeks in five. The concentration of voltage gated calcium channel-complex-reactive antibodies was 900 pM in one patient, and declined to 609 pM ∼ 3 months later. Measles-reactive IgG antibody avidity was high in AME patients born after vaccine coverage exceeded 50% (∼ 25 years earlier). AME patients had low CD4 (218/µl, p = 0.029) and CD8 (200/µl, p = 0.012) T-cell counts compared to controls. CONCLUSION Young adults presenting with AME in Vietnam reported a history of one prior measles immunization, and those aged <25 years had high measles-reactive IgG avidity indicative of prior vaccination. This suggests that one-dose measles immunization is not sufficient to prevent AME in young adults and reinforces the importance of maintaining high coverage with a two-dose measles immunization schedule. Treatment with corticosteroids and IVIG is common practice, and should be assessed in randomized clinical trials.
Collapse
Affiliation(s)
- Annette Fox
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, Ho Chi Minh City and Ha Noi, Viet Nam.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Pasquali JL, Martin T. Control of B cells expressing naturally occurring autoantibodies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 750:145-56. [PMID: 22903672 DOI: 10.1007/978-1-4614-3461-0_11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Naturally occurring autoantibodies (NAbs) are typically polyreactive, bind with low affinity to a discrete set of autoantigens and are encoded by variable region genes in germline configuration. They differ from disease-associated autoantibodies (autoAb), which are mostly monoreactive, somatically mutated and of high affinities. Structure-function studies have shown that polyreactivity of NAbs relies on the somatically generated complementarity determining region, CDR3, of the heavy chain. This finding suggested that NAb-producing B cells were positively selected from the pre-immune B-cell repertoire. The biological significance of this selection remains, however, unclear. Data originating mainly from transgenic mice have shown that mature NAb-producing B cells are frequently ignorant toward their antigen, possibly due to their low affinity, though active tolerance mechanisms are not excluded. An important issue is whether NAb-producing B cells constitute the pool from which pathologic auto Ab emerge after autoantigen-driven maturation. We summarize results obtained in mouse models, showing that some infectious agents are able to induce an autoantigen-driven activation of certain NAb-producing B cells. However direct proof that selection by autoantigen may lead to somatic hypermutation are still lacking. Other data tend to suggest that pathologic auto Abs may derive from non-autoimmune B cells that have diversified by somatic hypermutation of their variable region genes.
Collapse
Affiliation(s)
- Jean Louis Pasquali
- Clinical Immunology Department, National Referral Center for Systemic Autoimmune Diseases, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | | |
Collapse
|
32
|
Theise ND, Bodenheimer HC, Ferrell LD. Acute and chronic viral hepatitis. MACSWEEN'S PATHOLOGY OF THE LIVER 2012:361-401. [DOI: 10.1016/b978-0-7020-3398-8.00007-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
33
|
Stübgen JP. Immune-mediated myelitis associated with hepatitis virus infections. J Neuroimmunol 2011; 239:21-7. [PMID: 21945641 DOI: 10.1016/j.jneuroim.2011.09.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 07/24/2011] [Accepted: 09/02/2011] [Indexed: 12/14/2022]
Abstract
Virus-induced spinal cord damage results from a cytolytic effect on anterior horn cells or from predominantly cellular immune-mediated damage of long white matter tracts. Infection with the hepatitis virus group, most notably hepatitis C virus, has infrequently been associated with the occurrence of myelitis. The pathogenesis of hepatitis virus-associated myelitis has not been clarified: virus-induced autoimmunity (humoral or cell-mediated, possibly vasculitic) seems the most likely disease mechanism. Limited available information offers no evidence of direct hepatitis virus infection of the spinal cord. Virus neuropenetration may occur after virus-infected mononuclear cells penetrate the blood-brain barrier, but a true neurolytic effect has not been demonstrated. Attacks of acute myelitis usually respond favorably to immunomodulatory therapy. Antiviral therapy plays no confirmed role in the treatment of acute bouts of myelitis, but may limit the relapsing course of HCV-associated myelitis.
Collapse
Affiliation(s)
- Joerg-Patrick Stübgen
- Department of Neurology and Neuroscience, Cornell University Medical College/New York Presbyterian Hospital, 525 East 68th Street, New York, NY 10065-4885, USA.
| |
Collapse
|
34
|
Selmi C, Maria Papini A, Pugliese P, Claudia Alcaro M, Gershwin ME. Environmental pathways to autoimmune diseases: the cases of primary biliary cirrhosis and multiple sclerosis. Arch Med Sci 2011; 7:368-80. [PMID: 22295019 PMCID: PMC3258751 DOI: 10.5114/aoms.2011.23398] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 05/15/2011] [Accepted: 05/19/2011] [Indexed: 12/21/2022] Open
Abstract
The pathways leading to autoimmunity remain enigmatic despite numerous lines of experimental inquiry and epidemiological evidence. The mechanisms leading to the initiation and perpetuation of specific diseases such as primary biliary cirrhosis (PBC) or multiple sclerosis (MS) remain largely enigmatic, although it is established that a combination of genetic predisposition and environmental stimulation is required. The growing number of genome-wide association studies and the largely incomplete concordance for autoimmune diseases in monozygotic twins concur to support the role of the environment (including infectious agents and chemicals) in the breakdown of tolerance leading to autoimmunity through different mechanisms. In the present article we illustrate the current hypotheses related to an environmental impact on the onset of PBC and MS as two representative conditions investigated with complementary approaches. Indeed, while a role of post-translational antigen modifications has been proposed for MS, this field remain unexplored in PBC where, conversely, most evidence is gathered from geoepidemiology and experimental data on xenobiotics or infectious agents.
Collapse
Affiliation(s)
- Carlo Selmi
- Department of Medicine and Autoimmunity and Metabolism Unit, IRCCS Istituto Clinico Humanitas, Rozzano, Milan, Italy
| | | | | | | | | |
Collapse
|
35
|
Poole BD, Kivovich V, Gilbert L, Naides SJ. Parvovirus B19 nonstructural protein-induced damage of cellular DNA and resultant apoptosis. Int J Med Sci 2011; 8:88-96. [PMID: 21278893 PMCID: PMC3030141 DOI: 10.7150/ijms.8.88] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Accepted: 01/13/2011] [Indexed: 12/19/2022] Open
Abstract
Parvovirus B19 is a widespread virus with diverse clinical presentations. The viral nonstructural protein, NS1, binds to and cleaves the viral genome, and induces apoptosis when transfected into nonpermissive cells, such as hepatocytes. We hypothesized that the cytotoxicity of NS1 in such cells results from chromosomal DNA damage caused by the DNA-nicking and DNA-attaching activities of NS1. Upon testing this hypothesis, we found that NS1 covalently binds to cellular DNA and is modified by PARP, an enzyme involved in repairing single-stranded DNA nicks. We furthermore discovered that the DNA nick repair pathway initiated by poly(ADPribose)polymerase and the DNA repair pathways initiated by ATM/ATR are necessary for efficient apoptosis resulting from NS1 expression.
Collapse
Affiliation(s)
- Brian D Poole
- Huck Institute for Life Sciences, Pennsylvania State University College of Medicine/Milton S. Hershey Medical Center, Hershey, PA, USA
| | | | | | | |
Collapse
|
36
|
Sugalski JM, Rodriguez B, Moir S, Anthony DD. Peripheral blood B cell subset skewing is associated with altered cell cycling and intrinsic resistance to apoptosis and reflects a state of immune activation in chronic hepatitis C virus infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 185:3019-27. [PMID: 20656924 PMCID: PMC3805966 DOI: 10.4049/jimmunol.1000879] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chronic hepatitis C virus (HCV) infection is associated with B cell activation, although underlying mechanisms are unclear. To investigate B cell regulation during HCV infection, we measured bulk B cell CpG and Staphylococcus aureus Cowan-induced IgG Ab-secreting cell (ASC) frequency, HCV and tetanus-specific ASC frequency, BCR- and CD40L-dependent CD80/CD86 expression, and activation of memory CD4 cells. Immature transitional, naive, resting memory, mature activated, tissue-like memory, and plasma B cell subset frequencies, cell cycling, and intrinsic apoptosis were quantified. We observed intact or enhanced tetanus-specific and total IgG ASC frequency, serum IgG, BCR- and CD40L-dependent CD80/CD86 expression, and CD40L-dependent bulk B cell activation of memory CD4 cells in HCV infection. HCV-specific ASCs were observed in HCV-infected but not control subjects, although frequencies were lower compared with tetanus-specific cells. Immature transitional and mature activated B cell subset frequencies were increased in HCV-infected subjects, with immature transitional frequency associated with liver inflammation and serum B cell-activating factor. Mature activated B cells less commonly expressed Ki67, more commonly expressed Bcl2, and were more intrinsically resistant to apoptosis, whereas immature transitional B cells more commonly expressed Ki67, the latter associated with plasma HCV level. Taken together, these results indicate that in the setting of chronic HCV infection, a state of activation results in B cell subset skewing that is likely the result of alterations in homeostasis, cell cycling, and intrinsic resistance to apoptosis and that results in an overall intact or enhanced B cell response to BCR and CD40L.
Collapse
Affiliation(s)
- Julia M Sugalski
- Department of Medicine, Case Western Reserve University Center for AIDS Research and VA Medical Center, Cleveland, OH 44106, USA
| | | | | | | |
Collapse
|
37
|
Chen DY, Tzang BS, Chen YM, Lan JL, Tsai CC, Hsu TC. The association of anti-parvovirus B19-VP1 unique region antibodies with antiphospholipid antibodies in patients with antiphospholipid syndrome. Clin Chim Acta 2010; 411:1084-9. [DOI: 10.1016/j.cca.2010.04.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2010] [Revised: 04/02/2010] [Accepted: 04/03/2010] [Indexed: 11/28/2022]
|
38
|
Abstract
Autoimmunity has been a topic of intensive research for several decades, yet amazingly, no uniform hypothesis exists to explain the basis for the spectrum of autoantibody specificities seen in autoimmune diseases. It therefore seems appropriate to consider whether our current framework for understanding tolerance, and thus the mechanisms controlling the initiation and perpetuation of autoimmunity, may be faulty. Adapting the paradigm of Matzinger-the 'danger model', a case can be made for a perspective that appreciates the fundamental role of the tissues in controlling immune response, favouring a shift of focus in studies on the initiation of autoimmunity. Applying the elements of this model, I set forth a number of scenarios for how autoreactivity could emerge, with emphasis on the likely sources of the involved autoantigens and the functional basis of their appearance. The emerging picture is one in which disruption of tissue homeostasis takes centre stage, with the antigen-presenting cells as the key players.
Collapse
Affiliation(s)
- Anders A Tveita
- Department of Biochemistry, Institute of Medical Biology, University of Tromsø, N-9037 Tromsø, Norway.
| |
Collapse
|
39
|
Presence of autoimmune antibody in chikungunya infection. Case Rep Med 2009; 2009:840183. [PMID: 19997520 PMCID: PMC2787058 DOI: 10.1155/2009/840183] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Accepted: 10/28/2009] [Indexed: 11/17/2022] Open
Abstract
Chikungunya infection has recently re-emerged as an important arthropod-borne disease in Thailand. Recently, Southern Thailand was identified as a potentially endemic area for the chikungunya virus. Here, we report a case of severe musculoskeletal complication, presenting with muscle weakness and swelling of the limbs. During the investigation to exclude autoimmune muscular inflammation, high titers of antinuclear antibody were detected. This is the report of autoimmunity detection associated with an arbovirus infection. The symptoms can mimic autoimmune polymyositis disease, and the condition requires close monitoring before deciding to embark upon prolonged specific treatment with immunomodulators.
Collapse
|
40
|
Yeoman AD, Westbrook RH, Al-Chalabi T, Carey I, Heaton ND, Portmann BC, Heneghan MA. Diagnostic value and utility of the simplified International Autoimmune Hepatitis Group (IAIHG) criteria in acute and chronic liver disease. Hepatology 2009; 50:538-45. [PMID: 19575457 DOI: 10.1002/hep.23042] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
UNLABELLED Diagnostic criteria for autoimmune hepatitis (AIH) have been created and revised by the International Autoimmune Hepatitis Group (IAIHG). Simplified criteria have been created, but remain independently unvalidated. We report on the diagnostic accuracy of the simplified criteria in patients across a range of diagnoses, including a subset of patients presenting with fulminant liver failure who required liver transplant. Patients with AIH and non-AIH etiologies of liver disease were identified from dedicated patient databases. Parameters relevant to the simplified and 1999 IAIHG criteria were recorded, and sensitivity, specificity, and positive and negative predictive values for scores of >or=6 (probable AIH) and >or=7 (definite AIH) were calculated. A total of 549 patients with chronic liver disease were evaluated, (221 with AIH, 26 with variant syndromes, and 302 with non-AIH). For scores >or=6, sensitivity was 90%, and specificity was 98% with positive and negative predictive values of 97% and 92%, respectively. For scores >or=7; sensitivity was 70%, and specificity was 100% with positive and negative predictive values of 100% and 74%, respectively. Seven false positive diagnoses of AIH occurred, all with simplified scores of 6. Concordance with 1999 criteria was 90% for probable and 61% for definite AIH. The frequency of an overall diagnosis of AIH (probable or definite AIH) among the 70 patients with fulminant liver failure was 24% for simplified criteria and 40% for 1999 criteria, respectively. CONCLUSION The simplified criteria retain high specificity but exhibit lower sensitivity for scores of >or=7. The explanations for this are unclear but may relate to loss of such discriminating information as response to corticosteroids. Prospective evaluation of these criteria is required to corroborate these observations.
Collapse
Affiliation(s)
- Andrew D Yeoman
- Institute of Liver Studies, King's College Hospital NHS Foundation Trust, London, UK
| | | | | | | | | | | | | |
Collapse
|
41
|
Heit A, Gebhardt F, Lahl K, Neuenhahn M, Schmitz F, Anderl F, Wagner H, Sparwasser T, Busch DH, Kastenmüller K. Circumvention of regulatory CD4(+) T cell activity during cross-priming strongly enhances T cell-mediated immunity. Eur J Immunol 2008; 38:1585-97. [PMID: 18465771 DOI: 10.1002/eji.200737966] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Immunization with purified antigens is a safe and practical vaccination strategy but is generally unable to induce sustained CD8(+) T cell-mediated protection against intracellular pathogens. Most efforts to improve the CD8(+) T cell immunogenicity of these vaccines have focused on co-administration of adjuvant to support cross-presentation and dendritic cell maturation. In addition, it has been shown that CD4(+) T cell help during the priming phase contributes to the generation of protective CD8(+) memory T cells. In this report we demonstrate that the depletion of CD4(+) T cells paradoxically enhances long-lasting CD8-mediated protective immunity upon protein vaccination. Functional and genetic in vivo inactivation experiments attribute this enhancement primarily to MHC class II-restricted CD4(+) regulatory T cells (Treg), which appear to physiologically suppress the differentiation process towards long-living effector memory T cells. Since, in functional terms, this suppression by Treg largely exceeds the positive effects of conventional CD4(+) T cell help, even the absence of all CD4(+) T cells or lack of MHC class II-mediated interactions on priming dendritic cells result in enhanced CD8(+) T cell immunogenicity. These findings have important implications for the improvement of vaccines against intracellular pathogens or tumors, especially in patients with highly active Treg.
Collapse
Affiliation(s)
- Antje Heit
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University Munich, Munich, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Subacute cutaneous lupus erythematosus and interstitial myositis occurring with hepatitis B infection: response to antiviral therapy with lamivudine. J Clin Rheumatol 2008; 14:346-9. [PMID: 18664991 DOI: 10.1097/rhu.0b013e31817de0e7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Lupus erythematosus (LE) is a multisystem inflammatory disease resulting from an altered T- and B-cell response. Among various pathogenic factors, viral infections have been implicated in LE exacerbation. We describe a patient with acute onset of subacute cutaneous lupus erythematosus (SCLE) and interstitial myositis with mitochondrial changes in the context of an associated hepatitis B infection and the response of myositis to antiviral treatment with lamivudine. Viral vectors may play an important role in LE. A hitherto undescribed myositis with mitochondrial changes was associated with hepatitis B infection and SCLE and improved with antiviral treatment.
Collapse
|
43
|
Prevalence of disease-specific antinuclear antibodies in general population: estimates from annual physical examinations of residents of a small town over a 5-year period. Mod Rheumatol 2008; 18:153-60. [PMID: 18283522 DOI: 10.1007/s10165-008-0028-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2007] [Accepted: 11/02/2007] [Indexed: 10/22/2022]
Abstract
The aim of this study was to investigate the types and prevalence of disease-specific antinuclear antibodies (ANAs) and their relationship to rheumatic diseases in the general Japanese population. An immunofluorescence (IF) method was used for the first screening of ANA levels in serum samples obtained from 2181 residents of a small Japanese town. Individuals positive for IF-ANA were then further tested for disease-specific ANAs using eight enzyme immunoassays. Physical status and the presence of illness were determined by means of questionnaires and medical examinations. Based on the result of the IF-ANA assay, the rates of positive samples at 1:40 and 1:160 dilutions were 26.0 and 9.5%, respectively, with females have significantly higher positivity rates than males (P < 0.0001). Among 566 IF-ANA-positive individuals, 100 individuals were found to have 114 disease-specific ANAs. Anti-SSA/Ro, anti-centromere, and anti-U1RNP antibodies were detected in 58, 30, and 11 individuals, respectively, but anti-Sm, anti-Scl-70, and anti-Jo-1 antibodies were undetectable. Questionnaires and medical examinations revealed that among 60 disease-specific ANA-positive individuals that were available for testing, six had Sjögren's syndrome (SS), five were suspected of having SS, and five had rheumatoid arthritis. Surprisingly, 34 (57%) of the disease-specific ANA-positive individuals were clinically healthy. Anti-SSA/Ro, anti-centromere, and anti-U1RNP antibodies were quite frequent among clinically healthy Japanese subjects, although anti-Sm, anti-Scl-70, and anti-Jo-1 antibodies were not. Of the 60 individuals who tested positive for disease-specific ANAs, 30% (18/60) actually manifested systemic rheumatic diseases, while 50% showed no detectable signs or symptoms of rheumatic diseases.
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW To review the current literature and summarize the main principles found between viral infections and the subsequent production of autoantibodies. RECENT FINDINGS We concentrate on recent findings involving three viral agents, one of which is Epstein-Barr virus, which has been associated with many autoimmune diseases and is classically considered to induce systemic lupus erythematosus. As we will discuss, this occurs through molecular mimicry between Epstein-Barr virus nuclear antigen 1 and lupus-specific antigens such as Ro, La or dsDNA, through induction of Toll-like receptor hypersensitivity by Epstein-Barr virus latent membrane protein 2A or by creating immortal B and T cells by loss of apoptosis. Hepatitis B virus was found to share amino acid sequences with different autoantigens. Tissue damage and the release of intracellular components is just another example of the autoantibody production caused by this virus. Cytomegalovirus has often been controversially associated with several autoimmune diseases and, although is the least understood viral infection of the three, appears to be somewhat suspicious. SUMMARY Understanding the infectious origin of autoimmune diseases is important as we aim to identify high-risk patients and disrupt this process with vaccines or other medications, ultimately delaying or even preventing the evolution of autoimmune diseases.
Collapse
|
45
|
Bernal W, Ma Y, Smith HM, Portmann B, Wendon J, Vergani D. The significance of autoantibodies and immunoglobulins in acute liver failure: a cohort study. J Hepatol 2007; 47:664-70. [PMID: 17602781 DOI: 10.1016/j.jhep.2007.05.011] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2006] [Revised: 05/02/2007] [Accepted: 05/07/2007] [Indexed: 12/16/2022]
Abstract
BACKGROUND/AIMS The cause of cryptogenic ('seronegative') acute liver failure (ALF) is unknown; the presence of non-organ specific autoantibodies (NOSA) may indicate an autoimmune pathogenesis. We investigated the prevalence of autoimmune features including antibodies to soluble liver antigen (anti-SLA) determined by Radioligand assay (RLA) in ALF patients. METHODS The prevalence and relationship to the immunoglobulin (Ig) response of NOSA and anti-SLA were determined in 73 ALF patients of different causes. Patients were scored using the revised system for diagnosis of autoimmune hepatitis (AIH). RESULTS Autoantibodies were present in 23 (32%) patients; 16 had anti-SLA, 6 ANA, 4 ASM and 1 AMA. They were absent in paracetamol-related disease and present in 23 of 53 (43%) of non-paracetamol (NP) cases (p<0.0003). Two of 16 (13%) with NP drug-induced (NPDI), 9 of 21 (43%) viral and 5 of 16 (31%) cryptogenic cases had anti-SLA. NOSA and anti-SLA were not associated with liver injury severity or outcome. IgM was higher in those with viral disease (p<0.00001). AIH scores classified 50% of cryptogenic cases as 'probable autoimmune hepatitis'. CONCLUSIONS Autoantibodies are common in ALF; cryptogenic cases have features suggestive of an autoimmune pathogenesis. IgM is elevated in viral cases but infrequently in ALF of other causes.
Collapse
Affiliation(s)
- William Bernal
- Institute of Liver Studies, King's College London School of Medicine, King's College Hospital, Denmark Hill, London SE5 9RS, UK
| | | | | | | | | | | |
Collapse
|
46
|
Abstract
Kawasaki Disease is a small-to-medium-vessel vasculitis that preferentially affects children. Kawasaki Disease can occur in adults, but the presentation may differ from that observed in children. Typical findings in both adults and children include fever, conjunctivitis, pharyngitis, and skin erythema progressing to a desquamating rash on the palms and soles. Adults more frequently present with cervical adenopathy (93% of adults vs. 15% of children), hepatitis (65% vs. 10%), and arthralgia (61% vs. 24-38%). In contrast, adults are less frequently affected by meningitis (10% vs. 34%), thrombocytosis (55% vs. 100%), and coronary artery aneurysms (5% vs. 18-25%). We report a case of acute Kawasaki Disease in a 24-year-old man who presented with rash, fever, and arthritis. He was successfully treated with high-dose aspirin and intravenous immunoglobulin (IVIG). Our case highlights the importance of considering Kawasaki Disease in adults presenting with symptoms commonly encountered in a general medical practice.
Collapse
Affiliation(s)
| | | | - Laura Zakowski
- University of Wisconsin, Madison, Wisconsin USA
- 1190 Health Sciences Learning Center, 700 Highland Avenue, Madison, Wisconsin 53705 USA
| |
Collapse
|
47
|
Jellison ER, Guay HM, Szomolanyi-Tsuda E, Welsh RM. Dynamics and magnitude of virus-induced polyclonal B cell activation mediated by BCR-independent presentation of viral antigen. Eur J Immunol 2007; 37:119-28. [PMID: 17163452 DOI: 10.1002/eji.200636516] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Hypergammaglobulinemia and production of autoantibodies occur during many viral infections, and studies have suggested that viral antigen-presenting B cells may become polyclonally activated by CD4 T cells in vivo in the absence of viral engagement of the BCR. However, we have reported that CD4 cells in lymphocytic choriomengitis virus (LCMV)-infected mice kill adoptively transferred B cells coated with LCMV class II peptides. We report here that most of the surviving naïve B cells presenting class II MHC peptides undergo an extensive differentiation process involving both proliferation and secretion of antibodies. Both events require CD4 cells and CD40/CD40L interactions but not MyD88-dependent signaling within the B cells. B cells taken from immunologically tolerant donor LCMV-carrier mice with high LCMV antigen load became activated following adoptive transfer into LCMV-infected hosts, suggesting that B cells present sufficient antigen for this process during a viral infection. No division or activation of B cells was detected at all in virus-infected hosts in the absence of cognate CD4 T cells and class II antigen. This approach, therefore, formally demonstrates and quantifies a virus-induced polyclonal proliferation and differentiation of B cells, which, due to their high proportion, would mostly have BCR not specific for the virus.
Collapse
Affiliation(s)
- Evan R Jellison
- Department of Pathology, Program in Immunology and Virology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | | | | | | |
Collapse
|
48
|
Hazzan R, Mukamel M, Yacobovich J, Yaniv I, Tamary H. Risk factors for future development of systemic lupus erythematosus in children with idiopathic thrombocytopenic purpura. Pediatr Blood Cancer 2006; 47:657-9. [PMID: 16933242 DOI: 10.1002/pbc.20970] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The risk of later development of systemic lupus erythematosus (SLE) in childhood idiopathic thrombocytopenic purpura (ITP) is currently unknown. We retrospectively evaluated the incidence and risk factors of this complication in 222 children with ITP who were followed for a mean of 4.2+/-4.9 years. During that time 3.6% of the children (8/222) developed SLE; all were females with positive anti-nuclear antibody (ANA), older (12.7+/-3.6 vs. 6.4+/-4.3 years old; P<0.01), and were more likely to have chronic ITP (87.5 vs. 46%; P=0.02), and had high titers of ANA. The majority of children with ITP who had a positive ANA (14/22, 64%) did not develop SLE.
Collapse
MESH Headings
- Antibodies, Antinuclear/blood
- Child
- Chronic Disease
- Comorbidity
- Disease Progression
- Female
- Follow-Up Studies
- Humans
- Incidence
- Israel/epidemiology
- Lupus Erythematosus, Systemic/blood
- Lupus Erythematosus, Systemic/diagnosis
- Lupus Erythematosus, Systemic/epidemiology
- Male
- Purpura, Thrombocytopenic, Idiopathic/blood
- Purpura, Thrombocytopenic, Idiopathic/diagnosis
- Purpura, Thrombocytopenic, Idiopathic/epidemiology
- Retrospective Studies
- Risk Factors
- Sex Distribution
Collapse
Affiliation(s)
- Rawi Hazzan
- Internal Medicine D, Rabin Medical Center, Schneider Children's Medical Center of Israel, Petah Tikva, and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | |
Collapse
|
49
|
Somers EC, Thomas SL, Smeeth L, Hall AJ. Autoimmune diseases co-occurring within individuals and within families: a systematic review. Epidemiology 2006; 17:202-17. [PMID: 16477262 DOI: 10.1097/01.ede.0000193605.93416.df] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Autoimmune diseases have been observed to coexist both within individuals and within families. It is unclear whether clinical reports of comorbid autoimmune diseases represent chance findings or true associations. This systematic review evaluates the current level of evidence on the coexistence of selected autoimmune diseases within individuals and families. We reviewed the associations among 4 TH1-associated autoimmune diseases: insulin-dependent diabetes mellitus, autoimmune (Hashimoto) thyroiditis, rheumatoid arthritis, and multiple sclerosis. METHODS Studies quantifying the coexistence between the selected diseases, published through March 2004, were identified from Medline and Embase searches. Study eligibility was determined on the basis of preestablished criteria, and relevant data were extracted according to a fixed protocol. We determined the prevalence of comorbid autoimmune disease according to index disease and then compiled summary statistics. Heterogeneity among studies was assessed by exact likelihood ratio tests and Monte Carlo inference. RESULTS We found 54 studies that met the eligibility criteria. Of these, 52 studies examined the coexistence of disease within individuals and 9 studies examined within-family associations. The majority of studies were uncontrolled and did not account for confounding factors. There was substantial evidence for heterogeneity among studies. Although inconclusive, the data appear to support an increased prevalence of autoimmune thyroiditis among patients with rheumatoid arthritis and those with insulin-dependent diabetes mellitus, and an inverse association between rheumatoid arthritis and multiple sclerosis. CONCLUSION Although the available evidence does not permit firm conclusions regarding comorbidities among the selected autoimmune diseases, results are sufficiently suggestive to warrant further study.
Collapse
Affiliation(s)
- Emily C Somers
- Department of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK.
| | | | | | | |
Collapse
|
50
|
Hsu TC, Tsay GJ, Chen TY, Liu YC, Tzang BS. Anti-PCNA autoantibodies preferentially recognize C-terminal of PCNA in patients with chronic hepatitis B virus infection. Clin Exp Immunol 2006; 144:110-6. [PMID: 16542372 PMCID: PMC1809633 DOI: 10.1111/j.1365-2249.2006.03046.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2006] [Indexed: 11/29/2022] Open
Abstract
We previously reported anti-PCNA autoantibodies in sera from patients with chronic HBV and HCV infection. To analyse the antigenic regions on proliferating cell nuclear antigen (PCNA) that confer autoantibody binding in patients with chronic hepatitis B (HBV) and C (HCV) infection, eight constructs including one wild type PCNA, one mutant type Y114A_PCNA and six C- or N-terminal PCNA truncations were generated. Sera from 185 patients with systemic lupus erythematosus (SLE), 178 with chronic HBV and 163 with chronic HCV infection, and 68 healthy individuals were examined for the presentation of anti-PCNA antibodies by enzyme linked immunosorbent assay (ELISA). By ELISA, anti-PCNA positive sera from patients with SLE, chronic HBV and HCV infection preferentially recognized the wild type PCNA more than the mutant type Y114A_PCNA (P < 0.05). The inhibition of binding by purified full-length rPCNA proteins with anti-PCNA positive sera was shown to exceed 70%. The inhibition of binding by purified truncated rPCNA proteins with sera from patients with chronic HBV and HCV infection and SLE was shown to confer dominant binding in T(L2) and T(L3). Moreover, the higher frequency of inhibition by using T(L3) was found in patients with chronic HBV infection. These data indicate that anti-PCNA autoantibodies preferentially recognize C-terminal of PCNA in patients with chronic HBV infection and may also provide advanced understanding between viral infection and autoimmunity for further study.
Collapse
Affiliation(s)
- T-C Hsu
- Institute of Immunology, Chung Shan Medical University, Taichung, Taiwan.
| | | | | | | | | |
Collapse
|