1
|
Karkar A, Nair P, Mukherjee M, Raghavan NG. Autoimmune response to COVID-19 characterised by eosinophilia, NETosis and EETosis. BMJ Case Rep 2025; 18:e260300. [PMID: 40132935 DOI: 10.1136/bcr-2024-260300] [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: 03/27/2025] Open
Abstract
A previously healthy man in his 50s was admitted with severe COVID-19 pneumonia requiring extracorporeal membrane oxygenation (ECMO) support. He was found to have persistent eosinophilia, with a peak level of 7.5×109/L. He had received multiple courses of steroids, including dexamethasone and methylprednisolone, with no improvement. On day 47, hydrocortisone was initiated for the treatment of septic shock. This resulted in the normalisation of the eosinophil count, which correlated with radiographic and clinical improvement. The patient was transitioned off ECMO and eventually weaned off mechanical ventilation. Studies conducted using the patient's serum identified circulating autoantibodies, which induced neutrophil extracellular traps (NETosis) and eosinophil extracellular traps (EETosis). A varying response of the eosinophils to different corticosteroids was observed in vivo that corresponded with the ex vivo experiments. This case highlights an unrecognised phenomenon of differential response to corticosteroids in severe COVID-19. Furthermore, this is the first reported case of EETosis in COVID-19.
Collapse
Affiliation(s)
- Aram Karkar
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Parameswaran Nair
- McMaster University, Hamilton, Ontario, Canada
- Firestone Institute for Respiratory Health, Hamiltn, Ontario, Canada
| | - Manali Mukherjee
- McMaster University, Hamilton, Ontario, Canada
- Firestone Institute for Respiratory Health, Hamiltn, Ontario, Canada
| | - Natya G Raghavan
- McMaster University, Hamilton, Ontario, Canada
- Firestone Institute for Respiratory Health, Hamiltn, Ontario, Canada
| |
Collapse
|
2
|
Galipeau Y, Castonguay N, McCluskie PS, Sonoda MT, Keeshan A, Collins E, Arnold C, Pelchat M, Burns K, Cooper C, Langlois M. Autoantibodies targeting angiotensin-converting enzyme 2 are prevalent and not induced by SARS-CoV-2 infection. FASEB J 2025; 39:e70390. [PMID: 39950298 PMCID: PMC11826374 DOI: 10.1096/fj.202402694r] [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: 11/04/2024] [Revised: 01/07/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025]
Abstract
Clinical outcomes resulting from SARS-CoV-2 infection vary widely, ranging from asymptomatic cases to the development of mild to severe respiratory illness, and in some instances, chronic lingering disease and mortality. The underlying biological mechanisms driving this wide spectrum of pathogenicity among certain individuals and demographics remain elusive. Autoantibodies have emerged as potential contributors to the severity of COVID-19. Although preliminary reports have suggested the induction of antibodies targeting Angiotensin-Converting Enzyme II (ACE2) post-infection, this assertion lacks confirmation in large-scale studies. In this study, our objective is to comprehensively characterize and quantify the prevalence and expression levels of autoantibodies directed against ACE2 in a sizable cohort (n = 464). Our findings reveal that ACE2-reactive IgM antibodies are the most prevalent, with an overall seroprevalence of 18.8%, followed by IgG at 10.3% and IgA at 6.3%. Longitudinal analysis of individuals with multiple blood draws showed stable ACE2 IgG and IgA levels over time. Upon stratifying individuals based on molecular testing for SARS-CoV-2 or serological evidence of past infection, no significant differences were observed between groups. Functional assessment of ACE2 autoantibodies demonstrated that they are non-neutralizing and failed to inhibit spike-ACE2 interaction or affect the enzymatic activity of ACE2. Our results highlight that ACE2 autoantibodies are prevalent in the general population and were not induced by SARS-CoV-2 infection in our cohort. Notably, we found no substantiated evidence supporting a direct role for ACE2 autoantibodies in SARS-CoV-2 pathogenesis.
Collapse
Affiliation(s)
- Yannick Galipeau
- Department of Biochemistry, Microbiology & Immunology, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - Nicolas Castonguay
- Department of Biochemistry, Microbiology & Immunology, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - Pauline S. McCluskie
- Department of Biochemistry, Microbiology & Immunology, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | | | - Alexa Keeshan
- School of Epidemiology and Public Health, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
- Clinical EpidemiologyOttawa Hospital Research InstituteOttawaOntarioCanada
| | - Erin Collins
- School of Epidemiology and Public Health, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
- Clinical EpidemiologyOttawa Hospital Research InstituteOttawaOntarioCanada
| | - Corey Arnold
- Department of Biochemistry, Microbiology & Immunology, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - Martin Pelchat
- Department of Biochemistry, Microbiology & Immunology, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
- Centre for Infection, Immunity and Inflammation (CI3)University of OttawaOttawaOntarioCanada
| | - Kevin Burns
- Division of Nephrology, Department of MedicineUniversity of OttawaOttawaOntarioCanada
- Ottawa Hospital Research InstituteOttawaOntarioCanada
| | - Curtis Cooper
- School of Epidemiology and Public Health, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
- Ottawa Hospital Research InstituteOttawaOntarioCanada
- Division of Infectious Diseases, Department of MedicineUniversity of OttawaOttawaOntarioCanada
| | - Marc‐André Langlois
- Department of Biochemistry, Microbiology & Immunology, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
- Centre for Infection, Immunity and Inflammation (CI3)University of OttawaOttawaOntarioCanada
| |
Collapse
|
3
|
Galipeau Y, Cooper C, Langlois MA. Autoantibodies in COVID-19: implications for disease severity and clinical outcomes. Front Immunol 2025; 15:1509289. [PMID: 39835117 PMCID: PMC11743527 DOI: 10.3389/fimmu.2024.1509289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
Few pathogens have historically been subjected to as intense scientific and clinical scrutiny as SARS-CoV-2. The genetic, immunological, and environmental factors influencing disease severity and post-infection clinical outcomes, known as correlates of immunity, remain largely undefined. Clinical outcomes of SARS-CoV-2 infection vary widely, ranging from asymptomatic cases to those with life-threatening COVID-19 symptoms. While most infected individuals return to their former health and fitness within a few weeks, some develop debilitating chronic symptoms, referred to as long-COVID. Autoimmune responses have been proposed as one of the factors influencing long-COVID and the severity of SARS-CoV-2 infection. The association between viral infections and autoimmune pathologies is not new. Viruses such as Epstein-Barr virus and cytomegalovirus, among others, have been shown to induce the production of autoantibodies and the onset of autoimmune conditions. Given the extensive literature on SARS-CoV-2, here we review current evidence on SARS-CoV-2-induced autoimmune pathologies, with a focus on autoantibodies. We closely examine mechanisms driving autoantibody production, particularly their connection with disease severity and long-COVID.
Collapse
Affiliation(s)
- Yannick Galipeau
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Curtis Cooper
- The Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Centre for Infection, Immunity and Inflammation (CI3), University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
4
|
Aghajani Mir M. Brain Fog: a Narrative Review of the Most Common Mysterious Cognitive Disorder in COVID-19. Mol Neurobiol 2024; 61:9915-9926. [PMID: 37874482 DOI: 10.1007/s12035-023-03715-y] [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: 09/06/2023] [Accepted: 10/14/2023] [Indexed: 10/25/2023]
Abstract
It has been more than three years since COVID-19 impacted the lives of millions of people, many of whom suffer from long-term effects known as long-haulers. Notwithstanding multiorgan complaints in long-haulers, signs and symptoms associated with cognitive characteristics commonly known as "brain fog" occur in COVID patients over 50, women, obesity, and asthma at excessive. Brain fog is a set of symptoms that include cognitive impairment, inability to concentrate and multitask, and short-term and long-term memory loss. Of course, brain fog contributes to high levels of anxiety and stress, necessitating an empathetic response to this group of COVID patients. Although the etiology of brain fog in COVID-19 is currently unknown, regarding the mechanisms of pathogenesis, the following hypotheses exist: activation of astrocytes and microglia to release pro-inflammatory cytokines, aggregation of tau protein, and COVID-19 entry in the brain can trigger an autoimmune reaction. There are currently no specific tests to detect brain fog or any specific cognitive rehabilitation methods. However, a healthy lifestyle can help reduce symptoms to some extent, and symptom-based clinical management is also well suited to minimize brain fog side effects in COVID-19 patients. Therefore, this review discusses mechanisms of SARS-CoV-2 pathogenesis that may contribute to brain fog, as well as some approaches to providing therapies that may help COVID-19 patients avoid annoying brain fog symptoms.
Collapse
Affiliation(s)
- Mahsa Aghajani Mir
- Deputy of Research and Technology, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
5
|
Bond NG, Shore KR, Engel EJ, Coonan EE, Al-Hasan F, Gbakie MA, Kamara FK, Kanneh L, Momoh M, Kanneh IM, Sandi JD, Elliott D, Ficenec SC, Smira AR, Fischer WA, Wohl DA, Robinson JE, Shaffer JG, Garry RF, Samuels RJ, Grant DS, Schieffelin JS. Ebola Virus-Specific Neutralizing Antibody Persists at High Levels in Survivors 2 Years After Resolution of Disease in a Sierra Leonean Cohort. J Infect Dis 2024; 230:e929-e937. [PMID: 38801652 PMCID: PMC11481455 DOI: 10.1093/infdis/jiae155] [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: 03/14/2023] [Indexed: 05/29/2024] Open
Abstract
Ebola virus (EBOV) infection results in Ebola virus disease (EVD), an often severe disease with a nonspecific presentation. Since its recognition, periodic outbreaks of EVD continue to occur in sub-Saharan Africa. The 2013-2016 West African EVD outbreak was the largest recorded, resulting in a substantial cohort of EVD survivors with persistent health complaints and variable immune responses. In this study, we characterize humoral immune responses in EVD survivors and their contacts in Eastern Sierra Leone. We found high levels of EBOV IgG in EVD survivors and lower yet substantial antibody levels in household contacts, suggesting subclinical transmission. Neutralizing antibody function was prevalent but variable in EVD survivors, raising questions about the durability of immune responses from natural infection with EBOV. Additionally, we found that certain discrete symptoms-ophthalmologic and auditory-are associated with EBOV IgG seropositivity, while an array of symptoms are associated with the presence of neutralizing antibody.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Mambu Momoh
- Kenema Government Hospital, Sierra Leone
- Eastern Technical University, Kenema, Sierra Leone
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Zhai X, Wu W, Zeng S, Miao Y. Advance in the mechanism and clinical research of myalgia in long COVID. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL IMMUNOLOGY 2024; 13:142-164. [PMID: 39310121 PMCID: PMC11411160 DOI: 10.62347/txvo6284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 07/18/2024] [Indexed: 09/25/2024]
Abstract
As severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to evolve, mortality rates of coronavirus disease 2019 (COVID-19) have significantly decreased. However, a variable proportion of patients exhibit persistent prolonged symptoms of COVID-19 infection (long COVID). This virus primarily attacks respiratory system, but numerous individuals complain persistent skeletal muscle pain or worsening pre-existing muscle pain post COVID-19, which severely affects the quality of life and recovery. Currently, there is limited research on the skeletal muscle pain in long COVID. In this brief review, we review potential pathological mechanisms of skeletal muscle pain in long COVID, and summarize the various auxiliary examinations and treatments for skeletal muscle pain in long COVID. We consider abnormal activation of inflammatory response, myopathy, and neurological damages as pivotal pathological mechanisms of skeletal muscle pain in long COVID. A comprehensive examination is significantly important in order to work out effective treatment plans and relieve skeletal muscle pain. So far, rehabilitation interventions for myalgia in long COVID contain but are not limited to drug, nutraceutical therapy, gut microbiome-targeted therapy, interventional therapy and strength training. Our study provides a potential mechanism reference for clinical researches, highlighting the importance of comprehensive approach and management of skeletal muscle pain in long COVID. The relief of skeletal muscle pain will accelerate rehabilitation process, improve activities of daily living and enhance the quality of life, promoting individuals return to society with profound significance.
Collapse
Affiliation(s)
- Xiuyun Zhai
- Department of Rehabilitation, Shanghai General Hospital, Shanghai Jiaotong UniversityNo. 100, Haining Road, Shanghai 200080, China
| | - Weijun Wu
- Department of Rehabilitation, Shanghai General Hospital, Shanghai Jiaotong UniversityNo. 100, Haining Road, Shanghai 200080, China
| | - Siliang Zeng
- Department of Rehabilitation Therapy, School of Health, Shanghai Normal University Tianhua CollegeNo. 1661, North Shengxin Road, Shanghai 201815, China
| | - Yun Miao
- Department of Rehabilitation, Shanghai General Hospital, Shanghai Jiaotong UniversityNo. 100, Haining Road, Shanghai 200080, China
- Department of Rehabilitation, School of International Medical Technology, Shanghai Sanda UniversityNo. 2727, Jinhai Road, Shanghai 201209, China
| |
Collapse
|
7
|
Ashmawy R, Hammouda EA, El-Maradny YA, Aboelsaad I, Hussein M, Uversky VN, Redwan EM. Interplay between Comorbidities and Long COVID: Challenges and Multidisciplinary Approaches. Biomolecules 2024; 14:835. [PMID: 39062549 PMCID: PMC11275036 DOI: 10.3390/biom14070835] [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: 05/02/2024] [Revised: 06/24/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Long COVID, a name often given to the persistent symptoms following acute SARS-CoV-2 infection, poses a multifaceted challenge for health. This review explores the intrinsic relationship between comorbidities and autoimmune responses in shaping the trajectory of long COVID. Autoantibodies have emerged as significant players in COVID-19 pathophysiology, with implications for disease severity and progression. Studies show immune dysregulation persisting months after infection, marked by activated innate immune cells and high cytokine levels. The presence of autoantibodies against various autoantigens suggests their potential as comorbid factors in long COVID. Additionally, the formation of immune complexes may lead to severe disease progression, highlighting the urgency for early detection and intervention. Furthermore, long COVID is highly linked to cardiovascular complications and neurological symptoms, posing challenges in diagnosis and management. Multidisciplinary approaches, including vaccination, tailored rehabilitation, and pharmacological interventions, are used for mitigating long COVID's burden. However, numerous challenges persist, from evolving diagnostic criteria to addressing the psychosocial impact and predicting disease outcomes. Leveraging AI-based applications holds promise in enhancing patient management and improving our understanding of long COVID. As research continues to unfold, unravelling the complexities of long COVID remains paramount for effective intervention and patient care.
Collapse
Affiliation(s)
- Rasha Ashmawy
- Clinical Research Administration, Directorate of Health Affairs, Ministry of Health and Population, Alexandria 21554, Egypt; (R.A.); (I.A.); (M.H.)
- Biomedical Informatics and Medical Statistics, Medical Research Institute, Alexandria University, Alexandria 21561, Egypt;
| | - Esraa Abdellatif Hammouda
- Biomedical Informatics and Medical Statistics, Medical Research Institute, Alexandria University, Alexandria 21561, Egypt;
- Clinical Research Department, El-Raml Pediatric Hospital, Ministry of Health and Population, Alexandria 21563, Egypt
| | - Yousra A. El-Maradny
- Pharmaceutical and Fermentation Industries Development Center, City of Scientific Research and Technological Applications (SRTA-City), New Borg EL-Arab 21934, Alexandria, Egypt;
- Microbiology and Immunology, Faculty of Pharmacy, Arab Academy for Science, Technology and Maritime Transport (AASTMT), El-Alamein Campus, Aswan 51718, Egypt
| | - Iman Aboelsaad
- Clinical Research Administration, Directorate of Health Affairs, Ministry of Health and Population, Alexandria 21554, Egypt; (R.A.); (I.A.); (M.H.)
| | - Mai Hussein
- Clinical Research Administration, Directorate of Health Affairs, Ministry of Health and Population, Alexandria 21554, Egypt; (R.A.); (I.A.); (M.H.)
| | - Vladimir N. Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Elrashdy M. Redwan
- Department of Biological Science, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications, New Borg EL-Arab 21934, Alexandria, Egypt
| |
Collapse
|
8
|
Netti GS, Soccio P, Catalano V, De Luca F, Khalid J, Camporeale V, Moriondo G, Papale M, Scioscia G, Corso G, Foschino MP, Lo Caputo S, Lacedonia D, Ranieri E. The Onset of Antinuclear Antibodies (ANAs) as a Potential Risk Factor for Mortality and Morbidity in COVID-19 Patients: A Single-Center Retrospective Study. Biomedicines 2024; 12:1306. [PMID: 38927513 PMCID: PMC11201662 DOI: 10.3390/biomedicines12061306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/06/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
The immune system's amplified response to SARS-CoV-2 may lead to the production of autoantibodies, but their specific impact on disease severity and outcome remains unclear. This study aims to assess if hospitalized COVID-19 patients face a worse prognosis based on ANA presence, even without autoimmune diseases. We performed a retrospective, single-center, observational cohort study, enrolling 638 COVID-19 patients hospitalized from April 2020 to March 2021 at Hospital "Policlinico Riuniti" of Foggia (Italy). COVID-19 patients with a positive ANA test exhibited a significantly lower 30-day survival rate (64.4% vs. 83.0%) and a higher likelihood of severe respiratory complications during hospitalization than those with negative ANA screening (35.4% vs. 17.0%) (p < 0.001). The association between poor prognosis and ANA status was identified by calculating the HALP score (Hemoglobin-Albumin-Lymphocyte-Platelet), which was lower in COVID-19 patients with a positive ANA test compared to ANA-negative patients (108.1 ± 7.4 vs. 218.6 ± 11.2 AU; p < 0.011). In detail, COVID-19 patients with a low HALP showed a lower 30-day survival rate (99.1% vs. 83.6% vs. 55.2% for high, medium, and low HALP, respectively; p < 0.001) and a higher incidence of adverse respiratory events compared to those with high and medium HALP (13.1% vs. 35.2% vs. 64.6% for high, medium, and low HALP, respectively; p < 0.001). In summary, ANA positivity in COVID-19 patients appears to be linked to a more aggressive disease phenotype with a reduced survival rate. Furthermore, we propose that the HALP score could serve as a valuable parameter to assess prognosis for COVID-19 patients.
Collapse
Affiliation(s)
- Giuseppe Stefano Netti
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (V.C.); (F.D.L.); (J.K.); (V.C.); (E.R.)
- Unit of Clinical Pathology, Department of Laboratory Diagnostics, University Hospital “Policlinico Riuniti”, 71122 Foggia, Italy; (M.P.); (G.C.)
| | - Piera Soccio
- Unit of Respiratory Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (P.S.); (G.M.); (G.S.); (M.P.F.); (D.L.)
| | - Valeria Catalano
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (V.C.); (F.D.L.); (J.K.); (V.C.); (E.R.)
| | - Federica De Luca
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (V.C.); (F.D.L.); (J.K.); (V.C.); (E.R.)
| | - Javeria Khalid
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (V.C.); (F.D.L.); (J.K.); (V.C.); (E.R.)
| | - Valentina Camporeale
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (V.C.); (F.D.L.); (J.K.); (V.C.); (E.R.)
| | - Giorgia Moriondo
- Unit of Respiratory Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (P.S.); (G.M.); (G.S.); (M.P.F.); (D.L.)
| | - Massimo Papale
- Unit of Clinical Pathology, Department of Laboratory Diagnostics, University Hospital “Policlinico Riuniti”, 71122 Foggia, Italy; (M.P.); (G.C.)
| | - Giulia Scioscia
- Unit of Respiratory Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (P.S.); (G.M.); (G.S.); (M.P.F.); (D.L.)
| | - Gaetano Corso
- Unit of Clinical Pathology, Department of Laboratory Diagnostics, University Hospital “Policlinico Riuniti”, 71122 Foggia, Italy; (M.P.); (G.C.)
- Clinical Biochemistry, Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Maria Pia Foschino
- Unit of Respiratory Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (P.S.); (G.M.); (G.S.); (M.P.F.); (D.L.)
| | - Sergio Lo Caputo
- Unit of Infectious Diseases, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy;
| | - Donato Lacedonia
- Unit of Respiratory Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (P.S.); (G.M.); (G.S.); (M.P.F.); (D.L.)
| | - Elena Ranieri
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (V.C.); (F.D.L.); (J.K.); (V.C.); (E.R.)
- Unit of Clinical Pathology, Department of Laboratory Diagnostics, University Hospital “Policlinico Riuniti”, 71122 Foggia, Italy; (M.P.); (G.C.)
| |
Collapse
|
9
|
Gusev E, Sarapultsev A. Exploring the Pathophysiology of Long COVID: The Central Role of Low-Grade Inflammation and Multisystem Involvement. Int J Mol Sci 2024; 25:6389. [PMID: 38928096 PMCID: PMC11204317 DOI: 10.3390/ijms25126389] [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: 05/28/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Long COVID (LC), also referred to as Post COVID-19 Condition, Post-Acute Sequelae of SARS-CoV-2 Infection (PASC), and other terms, represents a complex multisystem disease persisting after the acute phase of COVID-19. Characterized by a myriad of symptoms across different organ systems, LC presents significant diagnostic and management challenges. Central to the disorder is the role of low-grade inflammation, a non-classical inflammatory response that contributes to the chronicity and diversity of symptoms observed. This review explores the pathophysiological underpinnings of LC, emphasizing the importance of low-grade inflammation as a core component. By delineating the pathogenetic relationships and clinical manifestations of LC, this article highlights the necessity for an integrated approach that employs both personalized medicine and standardized protocols aimed at mitigating long-term consequences. The insights gained not only enhance our understanding of LC but also inform the development of therapeutic strategies that could be applicable to other chronic conditions with similar pathophysiological features.
Collapse
Affiliation(s)
| | - Alexey Sarapultsev
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia;
| |
Collapse
|
10
|
Li Q, Li J, Zhou M, Ge Y, Liu Z, Li T, Zhang L. Antiphospholipid antibody-related hepatic vasculitis in a juvenile after non-severe COVID-19: a case report and literature review. Front Immunol 2024; 15:1354349. [PMID: 38707895 PMCID: PMC11066154 DOI: 10.3389/fimmu.2024.1354349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/29/2024] [Indexed: 05/07/2024] Open
Abstract
Antiphospholipid antibodies (aPL) are both laboratory evidence and causative factors for a broad spectrum of clinical manifestations of antiphospholipid syndrome (APS), with thrombotic and obstetric events being the most prevalent. Despite the aPL-triggered vasculopathy nature of APS, vasculitic-like manifestations rarely exist in APS and mainly appear associated with other concurrent connective tissue diseases like systemic lupus erythematous. Several studies have characterized pulmonary capillaritis related to pathogenic aPL, suggesting vasculitis as a potential associated non-thrombotic manifestation. Here, we describe a 15-year-old girl who develops hepatic infarction in the presence of highly positive aPL, temporally related to prior non-severe COVID-19 infection. aPL-related hepatic vasculitis, which has not been reported before, contributes to liver ischemic necrosis. Immunosuppression therapy brings about favorable outcomes. Our case together with retrieved literature provides supportive evidence for aPL-related vasculitis, extending the spectrum of vascular changes raised by pathogenic aPL. Differentiation between thrombotic and vasculitic forms of vascular lesions is essential for appropriate therapeutic decision to include additional immunosuppression therapy. We also perform a systematic review to characterize the prevalence and clinical features of new-onset APS and APS relapses after COVID-19 for the first time, indicating the pathogenicity of aPL in a subset of COVID-19 patients.
Collapse
Affiliation(s)
- Qingyu Li
- Tsinghua Medicine, School of Medicine, Tsinghua University, Beijing, China
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jingya Li
- Tsinghua Medicine, School of Medicine, Tsinghua University, Beijing, China
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Menglan Zhou
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
| | - Ying Ge
- Department of Infectious Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Zhengyin Liu
- Department of Infectious Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Taisheng Li
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
- Department of Infectious Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Li Zhang
- Department of Infectious Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
11
|
Heil M. Self-DNA driven inflammation in COVID-19 and after mRNA-based vaccination: lessons for non-COVID-19 pathologies. Front Immunol 2024; 14:1259879. [PMID: 38439942 PMCID: PMC10910434 DOI: 10.3389/fimmu.2023.1259879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 12/26/2023] [Indexed: 03/06/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic triggered an unprecedented concentration of economic and research efforts to generate knowledge at unequalled speed on deregulated interferon type I signalling and nuclear factor kappa light chain enhancer in B-cells (NF-κB)-driven interleukin (IL)-1β, IL-6, IL-18 secretion causing cytokine storms. The translation of the knowledge on how the resulting systemic inflammation can lead to life-threatening complications into novel treatments and vaccine technologies is underway. Nevertheless, previously existing knowledge on the role of cytoplasmatic or circulating self-DNA as a pro-inflammatory damage-associated molecular pattern (DAMP) was largely ignored. Pathologies reported 'de novo' for patients infected with Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV)-2 to be outcomes of self-DNA-driven inflammation in fact had been linked earlier to self-DNA in different contexts, e.g., the infection with Human Immunodeficiency Virus (HIV)-1, sterile inflammation, and autoimmune diseases. I highlight particularly how synergies with other DAMPs can render immunogenic properties to normally non-immunogenic extracellular self-DNA, and I discuss the shared features of the gp41 unit of the HIV-1 envelope protein and the SARS-CoV 2 Spike protein that enable HIV-1 and SARS-CoV-2 to interact with cell or nuclear membranes, trigger syncytia formation, inflict damage to their host's DNA, and trigger inflammation - likely for their own benefit. These similarities motivate speculations that similar mechanisms to those driven by gp41 can explain how inflammatory self-DNA contributes to some of most frequent adverse events after vaccination with the BNT162b2 mRNA (Pfizer/BioNTech) or the mRNA-1273 (Moderna) vaccine, i.e., myocarditis, herpes zoster, rheumatoid arthritis, autoimmune nephritis or hepatitis, new-onset systemic lupus erythematosus, and flare-ups of psoriasis or lupus. The hope is to motivate a wider application of the lessons learned from the experiences with COVID-19 and the new mRNA vaccines to combat future non-COVID-19 diseases.
Collapse
Affiliation(s)
- Martin Heil
- Departamento de Ingeniería Genética, Laboratorio de Ecología de Plantas, Centro de Investigación y de Estudios Avanzados (CINVESTAV)-Unidad Irapuato, Irapuato, Mexico
| |
Collapse
|
12
|
Shen Y, Voigt A, Goranova L, Abed M, Kleiner DE, Maldonado JO, Beach M, Pelayo E, Chiorini JA, Craft WF, Ostrov DA, Ramiya V, Sukumaran S, Brown AN, Hanrahan KC, Tuanyok A, Warner BM, Nguyen CQ. Evidence of a Sjögren's disease-like phenotype following COVID-19 in mice and humans. JCI Insight 2023; 8:e166540. [PMID: 37676726 PMCID: PMC10807711 DOI: 10.1172/jci.insight.166540] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 09/05/2023] [Indexed: 09/09/2023] Open
Abstract
Sjögren's Disease (SjD) is a systemic autoimmune disease characterized by lymphocytic inflammation of the lacrimal and salivary glands (SG), dry eyes and mouth, and systemic symptoms. SARS-CoV-2 may trigger the development or progression of autoimmune diseases. To test this, we used a mouse model of SARS-CoV-2 infection and convalescent patients' blood and SG in order to understand the development of SjD-like autoimmunity after infection. First, SARS-CoV-2-infected human angiotensin-converting enzyme 2 (ACE2) transgenic mice exhibited decreased salivation, elevated antinuclear antibodies (ANA), and lymphocytic infiltration in the lacrimal and SG. The sera from patients with COVID-19 sera showed increased ANA (i.e., anti-SSA [Sjögren's-syndrome-related antigen A]/anti-Ro52 and anti-SSB [SS-antigen B]/anti-La). Male patients showed elevated anti-SSA compared with female patients, and female patients exhibited diverse ANA patterns. SG biopsies from convalescent COVID-19 patients were microscopically similar to SjD SG with focal lymphocytic infiltrates in 4 of 6 patients and 2 of 6 patients exhibiting focus scores of at least 2. Lastly, monoclonal antibodies produced in recovered patients blocked ACE2/spike interaction and cross-reacted with nuclear antigens. Our study shows a direct association between SARS-CoV-2 and SjD. Hallmark features of SjD-affected SGs were histologically indistinguishable from convalescent COVID-19 patients. The results implicate that SARS-CoV-2 could be an environmental trigger for SjD.
Collapse
Affiliation(s)
- Yiran Shen
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| | - Alexandria Voigt
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| | - Laura Goranova
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| | - Mehdi Abed
- Salivary Disorder Unit, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland, USA
| | - David E. Kleiner
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Jose O. Maldonado
- Salivary Disorder Unit, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland, USA
- AAV Biology Section, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland, USA
- Center for Oral Health Integration, HealthPartners Institute, Bloomington, MN, USA
| | - Margaret Beach
- Salivary Disorder Unit, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland, USA
| | - Eileen Pelayo
- Salivary Disorder Unit, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland, USA
| | - John A. Chiorini
- AAV Biology Section, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland, USA
| | - William F. Craft
- Department of Comparative, Diagnostic, and Population Medicine, College of Veterinary Medicine, and
| | - David A. Ostrov
- Department of Pathology, Immunology & Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Vijay Ramiya
- LifeSouth Community Blood Centers, Gainesville, Florida, USA
| | | | - Ashley N. Brown
- Institute for Therapeutic Innovation, Department of Medicine, University of Florida College of Medicine, Orlando, Florida, USA
| | - Kaley C. Hanrahan
- Institute for Therapeutic Innovation, Department of Medicine, University of Florida College of Medicine, Orlando, Florida, USA
| | - Apichai Tuanyok
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| | - Blake M. Warner
- Salivary Disorder Unit, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland, USA
| | - Cuong Q. Nguyen
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
- Department of Oral Biology, College of Dentistry and
- Center of Orphaned Autoimmune Diseases, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
13
|
Kozłowski P, Lulek M, Skwarek A, Śmiarowski M, Małecka-Giełdowska M, Ciepiela O. Mild-to-moderate COVID-19 does not predispose to the development of autoimmune rheumatic diseases or autoimmune-based thrombosis. Scand J Immunol 2023; 98:e13313. [PMID: 38441212 DOI: 10.1111/sji.13313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/16/2023] [Indexed: 03/07/2024]
Abstract
An infection with severe acute respiratory syndrome coronavirus (SARS-CoV-2) may have a significant impact on the human immune system. Interactions between the virus and defence mechanisms may promote the development of autoimmune processes which manifest as antinuclear antibody (ANA) synthesis. Since many different viruses are suspected to take part in the pathogenesis of different systemic autoimmune rheumatic diseases (SARDs), we examined whether coronavirus disease 2019 convalescents who suffer from mild to moderate disease have a higher risk of developing ANA and anti-β2-glicoprotein I IgG antibodies (β2 GPI). In a retrospective study, we examined 294 adults among volunteer blood donors divided into convalescents (N = 215) and healthy controls (N = 79). For ANA detection, we used line-blotting, a type of indirect immunofluorescence assay (IF), to determine antigenic specificity and ELISA for β2 GPI. We found a lower incidence of ANA in convalescents than in healthy controls, with the majority of these antibodies directed to antigens with no known clinical significance. Additionally, no participants were positive for β2 GPI in either group. Our results show that COVID-19 with mild to moderate symptoms in the generally healthy population does not induce the development of ANA or anti-β2 GPI antibodies for at least 6 months following the disease.
Collapse
Affiliation(s)
- Paweł Kozłowski
- Central Laboratory, University Clinical Centre of Medical University of Warsaw, Warsaw, Poland
| | - Michalina Lulek
- Students Scientific Group of Laboratory Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Agata Skwarek
- Students Scientific Group of Laboratory Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Marcin Śmiarowski
- Students Scientific Group of Laboratory Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Milena Małecka-Giełdowska
- Central Laboratory, University Clinical Centre of Medical University of Warsaw, Warsaw, Poland
- Department of Laboratory Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Olga Ciepiela
- Central Laboratory, University Clinical Centre of Medical University of Warsaw, Warsaw, Poland
- Department of Laboratory Medicine, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
14
|
Muyayalo KP, Gong GS, Kiyonga Aimeé K, Liao AH. Impaired immune response against SARS-CoV-2 infection is the major factor indirectly altering reproductive function in COVID-19 patients: a narrative review. HUM FERTIL 2023; 26:778-796. [PMID: 37811836 DOI: 10.1080/14647273.2023.2262757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/25/2023] [Indexed: 10/10/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease affecting multiple systems and organs, including the reproductive system. SARS-CoV-2, the virus that causes COVID-19, can damage reproductive organs through direct (angiotensin converting enzyme-2, ACE-2) and indirect mechanisms. The immune system plays an essential role in the homeostasis and function of the male and female reproductive systems. Therefore, an altered immune response related to infectious and inflammatory diseases can affect reproductive function and fertility in both males and females. This narrative review discussed the dysregulation of innate and adaptive systems induced by SARS-CoV-2 infection. We reviewed the evidence showing that this altered immune response in COVID-19 patients is the major indirect mechanism leading to adverse reproduction outcomes in these patients. We summarized studies reporting the long-term effect of SARS-CoV-2 infection on women's reproductive function and proposed the chronic inflammation and chronic autoimmunity characterizing long COVID as potential underlying mechanisms. Further studies are needed to clarify the role of autoimmunity and chronic inflammation (long COVID) in altered female reproduction function in COVID-19.
Collapse
Affiliation(s)
- Kahindo P Muyayalo
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
- Department of Obstetrics and Gynecology, University of Kinshasa, Kinshasa, D. R. Congo
| | - Guang-Shun Gong
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Kahindo Kiyonga Aimeé
- Department of Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, People's Republic of China
- Department of Tropical Medicine Infectious and Parasitic Diseases, University of Kinshasa, Kinshasa, D. R. Congo
| | - Ai-Hua Liao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| |
Collapse
|
15
|
Carletti P, Shah A, Bair C, Curran C, Mai A, Patel R, Moorthy R, Villate N, Davis JL, Vitale AT, Shakoor A, Hassman L. The spectrum of COVID-19-associated chorioretinal vasculopathy. Am J Ophthalmol Case Rep 2023; 31:101857. [PMID: 37255549 PMCID: PMC10193817 DOI: 10.1016/j.ajoc.2023.101857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/08/2023] [Accepted: 05/02/2023] [Indexed: 06/01/2023] Open
Abstract
Purpose Although conjunctivitis represents the most common ocular manifestation of COVID-19 infection, sight-threatening retinal involvement has been reported. Herein, we report and characterize with multimodal retinal imaging 5 cases of acute vision loss secondary to presumed chorioretinal vasculopathy temporally associated with COVID-19 infection with varying severity, visual morbidity, and treatment response, and review the available literature on the association between COVID-19 infection and retinal microvascular changes. Design Observational case series and literature review. Methods Multicenter case series of 5 patients who presented to academic centers and private offices with acute vision loss temporally associated with COVID-19 infection. A review of the literature was conducted using online databases. Results 10 eyes of 5 patients, 3 men and 2 women, with a mean age of 30.8 years (median 33, range 16-44) were described. All patients had a recently preceding episode of COVID-19, with symptomatology ranging from mild infection to life-threatening encephalopathy. Treatment for their retinal disease included topical, oral, intravitreal, and intravenous steroids, steroid-sparing immunosuppression, retinal photocoagulation, antivirals, and antiplatelet and anticoagulant agents. Treatment response and visual recovery ranged from complete recovery of baseline acuity to permanent vision loss and need for chronic immunosuppression. Conclusions and Importance Clinicians should be mindful of the potential for vision-threatening retinal involvement after COVID-19 infection. If found, treatment with both anti-inflammatory therapy and anticoagulation should be considered, in addition to close monitoring, as some patients with this spectrum of disease may require chronic immune suppression and/or anti-VEGF therapy.
Collapse
Affiliation(s)
- Piero Carletti
- Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Aaditya Shah
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Christopher Bair
- John A. Moran Eye Center, University of Utah Health, Salt Lake City, UT, United States
| | | | - Anthony Mai
- John A. Moran Eye Center, University of Utah Health, Salt Lake City, UT, United States
| | - Rachel Patel
- John A. Moran Eye Center, University of Utah Health, Salt Lake City, UT, United States
| | - Ramana Moorthy
- Associated Vitreoretinal and Uveitis Consultants, Indianapolis, IN, USA
| | - Natalia Villate
- Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
- Fort Lauderdale Eye Institute, Fort Lauderdale, FL, USA
| | - Janet L Davis
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Albert T Vitale
- John A. Moran Eye Center, University of Utah Health, Salt Lake City, UT, United States
| | - Akbar Shakoor
- John A. Moran Eye Center, University of Utah Health, Salt Lake City, UT, United States
| | - Lynn Hassman
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
16
|
Oda JM, den Hartigh AB, Jackson SM, Tronco AR, Fink SL. The unfolded protein response components IRE1α and XBP1 promote human coronavirus infection. mBio 2023; 14:e0054023. [PMID: 37306512 PMCID: PMC10470493 DOI: 10.1128/mbio.00540-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 04/20/2023] [Indexed: 06/13/2023] Open
Abstract
The cellular processes that support human coronavirus replication and contribute to the pathogenesis of severe disease remain incompletely understood. Many viruses, including coronaviruses, cause endoplasmic reticulum (ER) stress during infection. IRE1α is a component of the cellular response to ER stress that initiates non-conventional splicing of XBP1 mRNA. Spliced XBP1 encodes a transcription factor that induces the expression of ER-related targets. Activation of the IRE1α-XBP1 pathway occurs in association with risk factors for severe human coronavirus infection. In this study, we found that the human coronaviruses HCoV-OC43 (human coronavirus OC43) and SARS-CoV-2 (severe acute respiratory syndrome coronavirus-2) both robustly activate the IRE1α-XBP1 branch of the unfolded protein response in cultured cells. Using IRE1α nuclease inhibitors and genetic knockdown of IRE1α and XBP1, we found that these host factors are required for optimal replication of both viruses. Our data suggest that IRE1α supports infection downstream of initial viral attachment and entry. In addition, we found that ER stress-inducing conditions are sufficient to enhance human coronavirus replication. Furthermore, we found markedly increased XBP1 in circulation in human patients with severe coronavirus disease 2019 (COVID-19). Together, these results demonstrate the importance of IRE1α and XBP1 for human coronavirus infection. IMPORTANCE There is a critical need to understand the cellular processes co-opted during human coronavirus replication, with an emphasis on identifying mechanisms underlying severe disease and potential therapeutic targets. Here, we demonstrate that the host proteins IRE1α and XBP1 are required for robust infection by the human coronaviruses, SARS-CoV-2 and HCoV-OC43. IRE1α and XBP1 participate in the cellular response to ER stress and are activated during conditions that predispose to severe COVID-19. We found enhanced viral replication with exogenous IRE1α activation, and evidence that this pathway is activated in humans during severe COVID-19. Together, these results demonstrate the importance of IRE1α and XBP1 for human coronavirus infection.
Collapse
Affiliation(s)
- Jessica M. Oda
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Andreas B. den Hartigh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Shoen M. Jackson
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Ana R. Tronco
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Susan L. Fink
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
17
|
Kouranloo K, Dey M, Elwell H, Nune A. A systematic review of the incidence, management and prognosis of new-onset autoimmune connective tissue diseases after COVID-19. Rheumatol Int 2023; 43:1221-1243. [PMID: 36786873 PMCID: PMC9927056 DOI: 10.1007/s00296-023-05283-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 01/30/2023] [Indexed: 02/15/2023]
Abstract
A literature review on new-onset autoimmune connective tissue diseases (ACTDs) following COVID-19 is lacking. We evaluated potential associations between COVID-19 and the development of new-onset ACTDs. The "population" was adults with disease terms for ACTDs, including systemic lupus erythematosus (SLE), Sjogren's syndrome, systemic sclerosis (SSc), idiopathic inflammatory myositis (IIM), anti-synthetase syndrome, mixed CTD and undifferentiated CTD, and "intervention" as COVID-19 and related terms. Databases were searched for English-language articles published until September 2022. We identified 2236 articles with 28 ultimately included. Of the 28 included patients, 64.3% were female, with a mean age was 51.1 years. The USA reported the most cases (9/28). ACTD diagnoses comprised: 11 (39.3%) IIM (including four dermatomyositis); 7 (25%) SLE; four (14.3%) anti-synthetase syndrome; four (14.3%) SSc; two (7.1%) other ACTD (one lupus/MCTD overlap). Of eight, four (14.3%) patients (including that with lupus/MCTD) had lupus nephritis. The average time from COVID-19 to ACTD diagnosis was 23.7 days. A third of patients were admitted to critical care, one for treatment of haemophagocytic lymphohistiocytosis in SLE (14 sessions of plasmapheresis, rituximab and intravenous corticosteroids) and nine due to COVID-19. 80% of patients went into remission of ACTD following treatment, while three (10%) patients died-one due to macrophage activation syndrome with anti-synthetase syndrome and two from unreported causes. Our results suggest a potential association between COVID-19 and new-onset ACTDs, notably in young females, reflecting more comprehensive CTD epidemiology. The most common diagnosis in our cohort was IIM. The aetiology and mechanisms by which ACTDs emerge following COVID-19 remain unknown and require further research.
Collapse
Affiliation(s)
- Koushan Kouranloo
- School of Medicine, University of Liverpool, Ashon St., Liverpool, L69 3GE, UK.
- Royal Liverpool University NHS Foundation Trust, Prescot St., Liverpool, L7 8XP, UK.
| | - Mrinalini Dey
- Department of Rheumatology, Queen Elizabeth Hospital, Stadium Rd., London, SE18 4QH, UK
- Institute of Life Health Sciences, University of Liverpool, Liverpool, L7 8TX, UK
| | - Helen Elwell
- BMA Library, BMA House, Tavistock Square, British Medical Association, London, WC1H 9JP, UK
| | - Arvind Nune
- Department of Rheumatology, Southport and Ormskirk NHS Foundation Trust, Southport, PR8 6PN, UK
| |
Collapse
|
18
|
Dima A, Popescu DN, Moroti R, Stoica E, State G, Negoi F, Berza IA, Parvu M. Antiphospholipid Antibodies Occurrence in Acute SARS-CoV-2 Infection without Overt Thrombosis. Biomedicines 2023; 11:biomedicines11051241. [PMID: 37238912 DOI: 10.3390/biomedicines11051241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/12/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
We sought to determine the prevalence of antiphospholipid antibodies (aPLs) and their correlation with COVID-19 severity (in terms of clinical and laboratory parameters) in patients without thrombotic events during the early phase of infection. This was a cross-sectional study with the inclusion of hospitalized COVID-19 patients from a single department during the COVID-19 pandemic (April 2020-May 2021). Previous known immune disease or thrombophilia along with long-term anticoagulation and patients with overt arterial or venous thrombosis during SARS-CoV-2 infection were excluded. In all cases, data on four criteria for aPL were collected, namely lupus anticoagulant (LA), IgM and IgG anticardiolipin antibodies (aCL), as well as IgG anti-β2 glycoprotein I antibodies (aβ2GPI). One hundred and seventy-nine COVID-19 patients were included, with a mean age of 59.6 (14.5) years and a sex ratio of 0.8 male: female. LA was positive in 41.9%, while it was strongly positive in 4.5%; aCL IgM was found in 9.5%, aCL IgG in 4.5%, and aβ2GPI IgG in 1.7% of the sera tested. Clinical correlation: LA was more frequently expressed in severe COVID-19 cases than in moderate or mild cases (p = 0.027). Laboratory correlation: In univariate analysis, LA levels were correlated with D-dimer (p = 0.016), aPTT (p = 0.001), ferritin (p = 0.012), C-reactive protein (CRP) (p = 0.027), lymphocyte (p = 0.040), and platelet (p < 0.001) counts. However, in the multivariate analysis, only the CRP levels correlated with LA positivity: OR (95% CI) 1.008 (1.001-1.016), p = 0.042. LA was the most common aPL identified in the acute phase of COVID-19 and was correlated with infection severity in patients without overt thrombosis.
Collapse
Affiliation(s)
- Alina Dima
- Department of Rheumatology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | | | - Ruxandra Moroti
- Department of Infectious Diseases, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Infectious Diseases, National Institute for Infectious Diseases Matei Bals, 021105 Bucharest, Romania
| | - Elisabeta Stoica
- Department of Infectious Diseases, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Georgiana State
- Department of Rheumatology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Florentina Negoi
- Department of Rheumatology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Ioana Adriana Berza
- Department of Rheumatology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Magda Parvu
- Department of Rheumatology, Colentina Clinical Hospital, 020125 Bucharest, Romania
| |
Collapse
|
19
|
Bossuyt X, Vulsteke JB, Van Elslande J, Boon L, Wuyts G, Willebrords S, Frans G, Geukens N, Carpentier S, Tejpar S, Wildiers H, Blockmans D, De Langhe E, Vermeersch P, Derua R. Antinuclear antibodies in individuals with COVID-19 reflect underlying disease: Identification of new autoantibodies in systemic sclerosis (CDK9) and malignancy (RNF20, RCC1, TRIP13). Autoimmun Rev 2023; 22:103288. [PMID: 36738952 PMCID: PMC9893804 DOI: 10.1016/j.autrev.2023.103288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
A high prevalence of antinuclear antibodies (ANA) in COVID-19 has been insinuated, but the nature of the target antigens is poorly understood. We studied ANA by indirect immunofluorescence in 229 individuals with COVID-19. The target antigens of high titer ANA (≥1:320) were determined by immunoprecipitation (IP) combined with liquid-chromatography-mass spectrometry (MS). High titer ANA (≥1:320) were found in 14 (6%) of the individuals with COVID-19. Of the 14 COVID-19 cases with high titer ANA, 6 had an underlying autoimmune disease and 5 a malignancy. IP-MS revealed known target antigens associated with autoimmune disease as well as novel autoantigens, including CDK9 (in systemic sclerosis) and RNF20, RCC1 and TRIP13 (in malignancy). The novel autoantigens were confirmed by IP-Western blotting. In conclusion, in depth analysis of the targets of high titer ANA revealed novel autoantigens in systemic sclerosis and in malignant disease.
Collapse
Affiliation(s)
- Xavier Bossuyt
- Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium; Clinical and Diagnostic Immunology, Department of Microbiology, Immunology and Transplantation, KU, Leuven, Belgium.
| | - Jean-Baptiste Vulsteke
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Belgium; Rheumatology, University Hospitals Leuven, Leuven, Belgium
| | - Jan Van Elslande
- Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Lise Boon
- Clinical and Diagnostic Immunology, Department of Microbiology, Immunology and Transplantation, KU, Leuven, Belgium
| | - Greet Wuyts
- Clinical and Diagnostic Immunology, Department of Microbiology, Immunology and Transplantation, KU, Leuven, Belgium
| | | | - Glynis Frans
- Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Nick Geukens
- KU Leuven, PharmAbs: The KU Leuven Antibody Center, Herestraat 49 Box 820, 3000 Leuven, Belgium
| | | | - Sabine Tejpar
- Laboratory of Molecular Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium; Department of Gastroenterology, University Hospitals Leuven, Belgium
| | - Hans Wildiers
- Department of General Medical Oncology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Daniel Blockmans
- General Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Ellen De Langhe
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Belgium; Rheumatology, University Hospitals Leuven, Belgium
| | - Pieter Vermeersch
- Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium; Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Rita Derua
- SyBioMa, KU Leuven, Belgium; Department of Cellular and Molecular Medicine, Laboratory of Protein Phosphorylation and Proteomics, KU Leuven, Belgium
| |
Collapse
|
20
|
Baiocchi GC, Vojdani A, Rosenberg AZ, Vojdani E, Halpert G, Ostrinski Y, Zyskind I, Filgueiras IS, Schimke LF, Marques AHC, Giil LM, Lavi YB, Silverberg JI, Zimmerman J, Hill DA, Thornton A, Kim M, De Vito R, Fonseca DLM, Plaça DR, Freire PP, Camara NOS, Calich VLG, Scheibenbogen C, Heidecke H, Lattin MT, Ochs HD, Riemekasten G, Amital H, Shoenfeld Y, Cabral-Marques O. Cross-sectional analysis reveals autoantibody signatures associated with COVID-19 severity. J Med Virol 2023; 95:e28538. [PMID: 36722456 DOI: 10.1002/jmv.28538] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 02/02/2023]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is associated with increased levels of autoantibodies targeting immunological proteins such as cytokines and chemokines. Reports further indicate that COVID-19 patients may develop a broad spectrum of autoimmune diseases due to reasons not fully understood. Even so, the landscape of autoantibodies induced by SARS-CoV-2 infection remains uncharted territory. To gain more insight, we carried out a comprehensive assessment of autoantibodies known to be linked to diverse autoimmune diseases observed in COVID-19 patients in a cohort of 231 individuals, of which 161 were COVID-19 patients (72 with mild, 61 moderate, and 28 with severe disease) and 70 were healthy controls. Dysregulated IgG and IgA autoantibody signatures, characterized mainly by elevated concentrations, occurred predominantly in patients with moderate or severe COVID-19 infection. Autoantibody levels often accompanied anti-SARS-CoV-2 antibody concentrations while stratifying COVID-19 severity as indicated by random forest and principal component analyses. Furthermore, while young versus elderly COVID-19 patients showed only slight differences in autoantibody levels, elderly patients with severe disease presented higher IgG autoantibody concentrations than young individuals with severe COVID-19. This work maps the intersection of COVID-19 and autoimmunity by demonstrating the dysregulation of multiple autoantibodies triggered during SARS-CoV-2 infection. Thus, this cross-sectional study suggests that SARS-CoV-2 infection induces autoantibody signatures associated with COVID-19 severity and several autoantibodies that can be used as biomarkers of COVID-19 severity, indicating autoantibodies as potential therapeutical targets for these patients.
Collapse
Affiliation(s)
- Gabriela C Baiocchi
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Aristo Vojdani
- Immunosciences Laboratory, Inc., Department of Immunology, Los Angeles, California, USA.,Cyrex Laboratories, Phoenix, Arizona, USA
| | - Avi Z Rosenberg
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Gilad Halpert
- Ariel University, Ariel, Israel.,Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel.,Saint Petersburg State University Russia, St Petersburg, Russia
| | - Yuri Ostrinski
- Ariel University, Ariel, Israel.,Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel.,Saint Petersburg State University Russia, St Petersburg, Russia
| | - Israel Zyskind
- Department of Pediatrics, NYU Langone Medical Center, New York, New York, USA.,Maimonides Medical Center, Brooklyn, New York, USA
| | - Igor S Filgueiras
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Lena F Schimke
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Alexandre H C Marques
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Lasse M Giil
- Department of Internal Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway
| | - Yael B Lavi
- Department of Chemistry Ben Gurion University Beer-Sheva, Beer-Sheva, Israel
| | - Jonathan I Silverberg
- Department of Dermatology, George Washington University School of Medicine and Health Sciences, Washington, USA
| | | | | | | | - Myungjin Kim
- Data Science Initiative at Brown University, Providence, Rhode Island, USA
| | - Roberta De Vito
- Department of Biostatistics and the Data Science Initiative at Brown University, Providence, Rhode Island, USA
| | - Dennyson L M Fonseca
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of Sao Paulo (USP), Sao Paulo, Brazil
| | - Desireé R Plaça
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, São Paulo, Brazil
| | - Paula P Freire
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Niels O S Camara
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Vera L G Calich
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Carmen Scheibenbogen
- Institute for Medical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Harald Heidecke
- CellTrend Gesellschaft mit beschränkter Haftung (GmbH), Luckenwalde, Germany
| | - Miriam T Lattin
- Department of Biology, Yeshiva University, Manhatten, New York, USA
| | - Hans D Ochs
- Department of Pediatrics, University of Washington School of Medicine, and Seattle Children's Research Institute, Seattle, Washington, USA
| | - Gabriela Riemekasten
- Department of Rheumatology, University Medical Center Schleswig-Holstein Campus Lübeck, Lübeck, Germany
| | - Howard Amital
- Ariel University, Ariel, Israel.,Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel.,Department of Medicine B, Sheba Medical Center, Tel Hashomer, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel.,Saint Petersburg State University Russia, St Petersburg, Russia
| | - Otavio Cabral-Marques
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of Sao Paulo (USP), Sao Paulo, Brazil.,Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, São Paulo, Brazil.,Department of Pharmacy and Postgraduate Program of Health and Science, Federal University of Rio Grande do Norte, Natal, Brazil.,Department of Medicine, Division of Molecular Medicine, University of São Paulo School of Medicine, Baltimore, USA.,Laboratory of Medical Investigation 29, University of São Paulo School of Medicine, São Paulo, Brazil
| |
Collapse
|
21
|
Moafi M, Ebrahimi MJ, Hatami F, Javandoust Gharehbagh F, Ahmadzadeh A, Emam MM, Rajaei A, Mansouri D, Alavi Darazam I. Lupus Anticoagulant Is Associated with Critical Cases and High Mortality in COVID-19: A Literature Review. TANAFFOS 2023; 22:53-60. [PMID: 37920325 PMCID: PMC10618584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 01/04/2023] [Indexed: 11/04/2023]
Abstract
Background In severe COVID-19 cases, a hypercoagulable state may occur. Antiphospholipid syndrome-related auto-antibodies (APSRAs) contribute to coagulopathy, but their role in COVID- 19 remains unclear. We aimed to investigate the prevalence of positive APSRAs and their effect on clinical outcomes in confirmed COVID-19 patients. Materials and Methods In this cross-sectional study, severe hospitalized COVID-19 cases were enrolled. Demographic and clinical data were obtained from the day of admission. APSRAs including IgG and/or IgM anticardiolipin (aCL) and anti-β2-glycoprotein1 (anti-β2GP1) as well as lupus anticoagulant (LAC) were measured. Results In this study, 54 severe COVID-19 cases with positive RT-PCR and chest CT scans were recruited. Positive APSRAs were found in 7 (12.9%) patients. Positive LAC was a more prevalent marker as compared to other tests (11.1%). The prevalence of positive aCL (IgM or IgG) and anti-ß2 GPI (IgM or IgG) was 1.8% (in an elderly woman). Lower oxygen saturation was found in the positive APSRAs group as opposed to the negative APSRAs group (70.3±9 vs. 84.8±9.7%). The mortality rate in the positive APSRAs group was significantly higher relative to the negative APSRAs group (83.3% vs. 27.1%; P-value: 0.01). Likewise, the mechanical ventilation requirement in the positive group was also higher (50% vs. 27.1%, P-value: 0.28). Conclusion This study indicated that LAC might be associated with critical cases and high mortality of COVID-19. Nonetheless, the mortality was not related to macrothrombotic incidence.
Collapse
Affiliation(s)
- Maral Moafi
- Department of Infectious Diseases and Tropical Medicine, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Ebrahimi
- Department of Infectious Diseases and Tropical Medicine, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Firouze Hatami
- Department of Infectious Diseases and Tropical Medicine, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farid Javandoust Gharehbagh
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arman Ahmadzadeh
- Rheumatology Department, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Mehdi Emam
- Rheumatology Department, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Rajaei
- Rheumatology Department, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Mansouri
- Division of Infectious Diseases and Clinical Immunology, National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Teheran, Iran
| | - Ilad Alavi Darazam
- Department of Infectious Diseases and Tropical Medicine, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Serrano M, Espinosa G, Serrano A, Cervera R. COVID-19 and the antiphospholipid syndrome. Autoimmun Rev 2022; 21:103206. [PMID: 36195247 PMCID: PMC9527199 DOI: 10.1016/j.autrev.2022.103206] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 09/28/2022] [Indexed: 01/08/2023]
Abstract
Coronavirus disease 2019 (COVID-19) has resulted in a global pandemic. Most COVID-19 patients are asymptomatic or have flu-like symptoms. However, around 15% of the patients may have severe disease, including unilateral or bilateral pneumonia with acute respiratory distress syndrome and progressive hypoxemia that may require mechanical ventilation assistance. A systemic inflammatory response syndrome occurs in the most severe forms of COVID-19, with multiorgan involvement which can be life threatening caused by a cytokine storm. Although what best characterizes COVID-19 are the manifestations of the respiratory system, it has been shown that it also acts at the cardiovascular level, producing coagulation abnormalities, which causes thrombotic events mainly in the arteries/arterioles, microcirculation and venous system, and potentially increased mortality risk. This multiorgan vascular disease overlaps with other known microangiopathies, such as thrombotic microangiopathy or paroxysmal nocturnal hemoglobinuria, where complement overactivation plays an important role in the pathophysiology of thrombosis. Furthermore, coagulopathy secondary to COVID-19 occurs in the context of an uncontrolled inflammatory response, reminiscent of APS, especially in its catastrophic form. This review summarizes the current knowledge regarding the relationship between COVID-19 and the APS.
Collapse
Affiliation(s)
- Manuel Serrano
- Department of Immunology, Healthcare Research Institute I+12, Hospital 12 de Octubre, Madrid, Spain
| | - Gerard Espinosa
- Department of Autoimmune Diseases, Hospital Clínic, Insititut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Antonio Serrano
- Department of Immunology, Healthcare Research Institute I+12, Hospital 12 de Octubre, Madrid, Spain
| | - Ricard Cervera
- Department of Autoimmune Diseases, Hospital Clínic, Insititut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Catalonia, Spain,Corresponding author at: Servei de Malalties Autoimmunes, Hospital Clínic, Villarroel, 170, 08036 Barcelona, Catalonia, Spain
| |
Collapse
|
23
|
Shen Y, Voigt A, Goranova L, Abed M, Kleiner DE, Maldonado JO, Beach M, Pelayo E, Chiorini JA, Craft WF, Ostrov DA, Ramiya V, Sukumaran S, Tuanyok A, Warner BM, Nguyen CQ. Evidence of a Sjögren's disease-like phenotype following COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.10.20.22281265. [PMID: 36324812 PMCID: PMC9628191 DOI: 10.1101/2022.10.20.22281265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Objectives Sjögren's Disease (SjD) is a chronic and systemic autoimmune disease characterized by lymphocytic infiltration and the development of dry eyes and dry mouth resulting from the secretory dysfunction of the exocrine glands. SARS-CoV-2 may trigger the development or progression of autoimmune diseases, as evidenced by increased autoantibodies in patients and the presentation of cardinal symptoms of SjD. The objective of the study was to determine whether SARS-CoV-2 induces the signature clinical symptoms of SjD. Methods The ACE2-transgenic mice were infected with SARS-CoV-2. SJD profiling was conducted. COVID-19 patients' sera were examined for autoantibodies. Clinical evaluations of convalescent COVID-19 subjects, including minor salivary gland (MSG) biopsies, were collected. Lastly, monoclonal antibodies generated from single B cells of patients were interrogated for ACE2/spike inhibition and nuclear antigens. Results Mice infected with the virus showed a decreased saliva flow rate, elevated antinuclear antibodies (ANAs) with anti-SSB/La, and lymphocyte infiltration in the lacrimal and salivary glands. Sera of COVID-19 patients showed an increase in ANA, anti-SSA/Ro52, and anti-SSB/La. The male patients showed elevated levels of anti-SSA/Ro52 compared to female patients, and female patients had more diverse ANA patterns. Minor salivary gland biopsies of convalescent COVID-19 subjects showed focal lymphocytic infiltrates in four of six subjects, and 2 of 6 subjects had focus scores >2. Lastly, we found monoclonal antibodies produced in recovered patients can both block ACE2/spike interaction and recognize nuclear antigens. Conclusion Overall, our study shows a direct association between SARS-CoV-2 and SjD. Hallmark features of SjD salivary glands were histologically indistinguishable from convalescent COVID-19 subjects. The results potentially implicate that SARS-CoV-2 could be an environmental trigger for SjD. Key Messages What is already known about this subject?SAR-CoV-2 has a tropism for the salivary glands. However, whether the virus can induce clinical phenotypes of Sjögren's disease is unknown.What does this study add?Mice infected with SAR-CoV-2 showed loss of secretory function, elevated autoantibodies, and lymphocyte infiltration in glands.COVID-19 patients showed an increase in autoantibodies. Monoclonal antibodies produced in recovered patients can block ACE2/spike interaction and recognize nuclear antigens.Minor salivary gland biopsies of some convalescent subjects showed focal lymphocytic infiltrates with focus scores.How might this impact on clinical practice or future developments?Our data provide strong evidence for the role of SARS-CoV-2 in inducing Sjögren's disease-like phenotypes.Our work has implications for how patients will be diagnosed and treated effectively.
Collapse
Affiliation(s)
- Yiran Shen
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| | - Alexandria Voigt
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| | - Laura Goranova
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| | - Mehdi Abed
- Salivary Disorder Unit, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland
| | - David E Kleiner
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Jose O Maldonado
- Salivary Disorder Unit, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland
- AAV Biology Section, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland, USA
| | - Margaret Beach
- Salivary Disorder Unit, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland
| | - Eileen Pelayo
- Salivary Disorder Unit, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland
| | - John A Chiorini
- AAV Biology Section, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland, USA
| | - William F Craft
- Department of Comparative, Diagnostic, and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| | - David A Ostrov
- Department of Pathology, Immunology & Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Vijay Ramiya
- LifeSouth Community Blood Centers, Gainesville Fl
| | | | - Apichai Tuanyok
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| | - Blake M Warner
- Salivary Disorder Unit, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland
| | - Cuong Q Nguyen
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
- Department of Oral Biology, College of Dentistry
- Center of Orphaned Autoimmune Diseases, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
24
|
Latorre D. Autoimmunity and SARS-CoV-2 infection: Unraveling the link in neurological disorders. Eur J Immunol 2022; 52:1561-1571. [PMID: 35833748 PMCID: PMC9350097 DOI: 10.1002/eji.202149475] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 05/14/2022] [Accepted: 07/12/2022] [Indexed: 12/14/2022]
Abstract
According to the World Health Organization, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has already infected more than 400 million people and caused over 5 million deaths globally. The infection is associated with a wide spectrum of clinical manifestations, ranging from no signs of illness to severe pathological complications that go beyond the typical respiratory symptoms. On this note, new-onset neurological and neuropsychiatric syndromes have been increasingly reported in a large fraction of COVID-19 patients, thus potentially representing a significant public health threat. Although the underlying pathophysiological mechanisms remain elusive, a growing body of evidence suggests that SARS-CoV-2 infection may trigger an autoimmune response, which could potentially contribute to the establishment and/or exacerbation of neurological disorders in COVID-19 patients. Shedding light on this aspect is urgently needed for the development of effective therapeutic intervention. This review highlights the current knowledge of the immune responses occurring in Neuro-COVID patients and discusses potential immune-mediated mechanisms by which SARS-CoV-2 infection may trigger neurological complications.
Collapse
|
25
|
Chen YC, Huang CP, Lin TM, Huang CY. Severe Digital Ischemia Associated with Upper Limb Arterial Thrombosis and Raynaud's Phenomenon in a Patient with Severe Acute Respiratory Syndrome Coronavirus 2 Infection. ACTA CARDIOLOGICA SINICA 2022; 38:650-652. [PMID: 36176367 PMCID: PMC9479058 DOI: 10.6515/acs.202209_38(5).20220330d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/30/2022] [Indexed: 01/24/2023]
Affiliation(s)
- Yen-Chou Chen
- Taipei Heart Institute, Taipei Medical University;
,
Division of Cardiology and Cardiovascular Research Center, Taipei Medical University Hospital
| | - Chien-Po Huang
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University
| | - Tzu-Min Lin
- Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Taipei Medical University Hospital;
,
Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei
| | - Chun-Yao Huang
- Taipei Heart Institute, Taipei Medical University;
,
Division of Cardiology and Cardiovascular Research Center, Taipei Medical University Hospital;
,
Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| |
Collapse
|
26
|
Butt A, Erkan D, Lee AI. COVID-19 and antiphospholipid antibodies. Best Pract Res Clin Haematol 2022; 35:101402. [PMID: 36494152 PMCID: PMC9568270 DOI: 10.1016/j.beha.2022.101402] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 12/14/2022]
Abstract
Antiphospholipid syndrome and the coagulopathy of COVID-19 share many pathophysiologic features, including endotheliopathy, hypercoagulability, and activation of platelets, complement pathways, and neutrophil extracellular traps, all acting in concert via a model of immunothrombosis. Antiphospholipid antibody production in COVID-19 is common, with 50% of COVID-19 patients being positive for lupus anticoagulant in some studies, and with non-Sapporo criteria antiphospholipid antibodies being prevalent as well. The biological significance of antiphospholipid antibodies in COVID-19 is uncertain, as such antibodies are usually transient, and studies examining clinical outcomes in COVID-19 patients with and without antiphospholipid antibodies have yielded conflicting results. In this review, we explore the biology of antiphospholipid antibodies in COVID-19 and other infections and discuss mechanisms of thrombogenesis in antiphospholipid syndrome and parallels with COVID-19 coagulopathy. In addition, we review the existing literature on safety of COVID-19 vaccination in patients with antiphospholipid antibodies and antiphospholipid syndrome.
Collapse
Affiliation(s)
- Ayesha Butt
- Section of Hematology, Department of Medicine, Yale School of Medicine, 333 Cedar St., New Haven, CT, 06520, USA.
| | - Doruk Erkan
- Barbara Volcker Center for Women and Rheumatic Diseases, Hospital for Special Surgery and Weill Cornell Medicine, 535 E. 70th St., 6th floor, New York, NY, 10021, USA.
| | - Alfred Ian Lee
- Section of Hematology, Department of Medicine, Yale School of Medicine, 333 Cedar St., New Haven, CT, 06520, USA.
| |
Collapse
|
27
|
Stjepanovic MI, Stojanovic MR, Stankovic S, Cvejic J, Dimic-Janjic S, Popevic S, Buha I, Belic S, Djurdjevic N, Stjepanovic MM, Jovanovic D, Stojkovic-Laloševic M, Soldatovic I, Bonaci-Nikolic B, Miskovic R. Autoimmune and immunoserological markers of COVID-19 pneumonia: Can they help in the assessment of disease severity. Front Med (Lausanne) 2022; 9:934270. [PMID: 36106319 PMCID: PMC9464912 DOI: 10.3389/fmed.2022.934270] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 08/08/2022] [Indexed: 12/04/2022] Open
Abstract
Background Immune dysregulation and associated inefficient anti-viral immunity during Coronavirus Disease 2019 (COVID-19) can cause tissue and organ damage which shares many similarities with pathogenetic processes in systemic autoimmune diseases. In this study, we investigate wide range autoimmune and immunoserological markers in hospitalized patients with COVID-19. Methods Study included 51 patients with confirmed Severe Acute Respiratory Syndrome Coronavirus 2 infection and hospitalized due to COVID-19 pneumonia. Wide spectrum autoantibodies associated with different autoimmune inflammatory rheumatic diseases were analyzed and correlated with clinical and laboratory features and pneumonia severity. Results Antinuclear antibodies (ANA) positivity was found in 19.6%, anti-cardiolipin IgG antibodies (aCL IgG) in 15.7%, and anti-cardiolipin IgM antibodies (aCL IgM) in 7.8% of patients. Positive atypical x anti-neutrophil cytoplasmic antibodies (xANCA) were detected in 10.0% (all negative for Proteinase 3 and Myeloperoxidase) and rheumatoid factor was found in 8.2% of patients. None of tested autoantibodies were associated with disease or pneumonia severity, except for aCL IgG being significantly associated with higher pneumonia severity index (p = 0.036). Patients with reduced total serum IgG were more likely to require non-invasive mechanical ventilation (NIMV) (p < 0.0001). Serum concentrations of IgG (p = 0.003) and IgA (p = 0.032) were significantly lower in this group of patients. Higher total serum IgA (p = 0.009) was associated with mortality, with no difference in serum IgG (p = 0.115) or IgM (p = 0.175). Lethal outcome was associated with lower complement C4 (p = 0.013), while there was no difference in complement C3 concentration (p = 0.135). Conclusion Increased autoimmune responses are present in moderate and severe COVID-19. Severe pneumonia is associated with the presence of aCL IgG, suggesting their role in disease pathogenesis. Evaluation of serum immunoglobulins and complement concentration could help assess the risk of non-invasive mechanical ventilation NIMV and poor outcome.
Collapse
Affiliation(s)
- Mihailo I. Stjepanovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic for Pulmonology, University Clinical Center of Serbia, Belgrade, Serbia
- *Correspondence: Mihailo I. Stjepanovic ;
| | - Maja R. Stojanovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic of Allergy and Immunology, University Clinical Center of Serbia, Belgrade, Serbia
| | - Sanja Stankovic
- Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Center for Medical Biochemistry, University Clinical Center of Serbia, Belgrade, Serbia
| | - Jelena Cvejic
- Clinic for Pulmonology, University Clinical Center of Serbia, Belgrade, Serbia
| | - Sanja Dimic-Janjic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic for Pulmonology, University Clinical Center of Serbia, Belgrade, Serbia
| | - Spasoje Popevic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic for Pulmonology, University Clinical Center of Serbia, Belgrade, Serbia
| | - Ivana Buha
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic for Pulmonology, University Clinical Center of Serbia, Belgrade, Serbia
| | - Slobodan Belic
- Clinic for Pulmonology, University Clinical Center of Serbia, Belgrade, Serbia
| | - Natasa Djurdjevic
- Clinic for Pulmonology, University Clinical Center of Serbia, Belgrade, Serbia
| | | | - Dragana Jovanovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic of Allergy and Immunology, University Clinical Center of Serbia, Belgrade, Serbia
| | - Milica Stojkovic-Laloševic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic of Gastroenterology and Hepatology, University Clinical Center of Serbia, Belgrade, Serbia
| | - Ivan Soldatovic
- Institute of Medical Statistics and Informatic, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Branka Bonaci-Nikolic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic of Allergy and Immunology, University Clinical Center of Serbia, Belgrade, Serbia
| | - Rada Miskovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Clinic of Allergy and Immunology, University Clinical Center of Serbia, Belgrade, Serbia
| |
Collapse
|
28
|
Torres-Ruiz J, Lomelín-Gascón J, Vargas-Castro AS, Lira-Luna J, Pérez-Fragoso A, Tapia-Conyer R, Nuñez-Aguirre M, Alcalá-Carmona B, Absalón-Aguilar A, Maravillas-Montero JL, Mejía-Domínguez NR, Núñez-Álvarez C, Rull-Gabayet M, Llorente L, Romero-Ramírez S, Sosa-Hernández VA, Cervantes-Díaz R, Juárez-Vega G, Meza-Sánchez DE, Martínez-Juárez LA, Morales-Juárez L, López-López LN, Negrete-Trujillo JA, Falcón-Lezama JA, Valdez-Vázquez RR, Gallardo-Rincón H, Gómez-Martín D. Clinical and immunological features associated to the development of a sustained immune humoral response in COVID-19 patients: Results from a cohort study. Front Immunol 2022; 13:943563. [PMID: 36045688 PMCID: PMC9421299 DOI: 10.3389/fimmu.2022.943563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/29/2022] [Indexed: 11/28/2022] Open
Abstract
Background Until now, most of the research addressing long-term humoral responses in coronavirus disease 2019 (COVID-19) had only evaluated the serum titers of anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) IgGs, without the assessment of the baseline antiviral clinical and immune profile, which is the aim of this study and may be the key factor leading to a broad and sustained antibody response. Methods We included 103 patients with COVID-19. When the patients sought medical attention (baseline), a blood sample was drawn to perform immunophenotype of lymphocytes by flow cytometry. The patients were assessed 15 days after baseline and then every month until the third month, followed by a last visit 6 months after recruitment. We evaluated the anti-SARS-COV-2 IgG at all time points, and the serum levels of cytokines, chemokines, anti-cellular (AC) antibodies and neutrophil extracellular traps were also assessed during the follow-up. The primary outcome of the study was the presence of a sustained immune humoral response, defined as an anti-SARS-CoV-2 IgG titer >4.99 arbitrary units/mL in at least two consecutive measures. We used generalized lineal models to assess the features associated with this outcome and to assess the effect of the changes in the cytokines and chemokines throughout time on the development of a sustained humoral immune response. Results At baseline the features associated to a sustained immune humoral response were the diagnosis of critical disease, absolute number of lymphocytes, serum IP-10, IL-4, IL-2, regulatory T cells, CD8+ T cells, and positive AC antibodies. Critical illness and the positivity of AC antibodies were associated with a sustained humoral immune response after 3 months, whilst critical illness and serum IL-13 were the explanatory variables after 6 months. Conclusion A sustained immune humoral response is strongly related to critical COVID-19, which is characterized by the presence of AC antibodies, quantitative abnormalities in the T cell compartment, and the serum cytokines and chemokines during acute infection and throughout time.
Collapse
Affiliation(s)
- Jiram Torres-Ruiz
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | - Ana Sofía Vargas-Castro
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jaquelin Lira-Luna
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina-Instituto Politécnico Nacional (IPN), Mexico City, Mexico
| | - Alfredo Pérez-Fragoso
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Laboratorio de Inmunoquimica 1, Posgrado en Ciencias Químicobiológicas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Roberto Tapia-Conyer
- Operative Solutions, Carlos Slim Foundation, Mexico City, Mexico
- Universidad Nacional Autónoma de México, Facultad de Medicina, Mexico City, Mexico
| | - Miroslava Nuñez-Aguirre
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Laboratorio de Inmunoquimica 1, Posgrado en Ciencias Químicobiológicas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Beatriz Alcalá-Carmona
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Laboratorio de Inmunoquimica 1, Posgrado en Ciencias Químicobiológicas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Abdiel Absalón-Aguilar
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Internal Medicine Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - José Luis Maravillas-Montero
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Nancy Raquel Mejía-Domínguez
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Carlos Núñez-Álvarez
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Marina Rull-Gabayet
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Luis Llorente
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Sandra Romero-Ramírez
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Victor Andrés Sosa-Hernández
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Rodrigo Cervantes-Díaz
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Guillermo Juárez-Vega
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - David Eduardo Meza-Sánchez
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Luis Alberto Martínez-Juárez
- Operative Solutions, Carlos Slim Foundation, Mexico City, Mexico
- Clinical Research, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Linda Morales-Juárez
- Operative Solutions, Carlos Slim Foundation, Mexico City, Mexico
- Temporary COVID-19 Hospital, Hipódromo de las Américas, Mexico City, Mexico
| | | | | | - Jorge Abelardo Falcón-Lezama
- Operative Solutions, Carlos Slim Foundation, Mexico City, Mexico
- Division of Health Sciences, Juárez Autonomous University of Tabasco, Tabasco, Mexico
| | | | - Héctor Gallardo-Rincón
- Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Guadalajara, Mexico
- *Correspondence: Diana Gómez-Martín, ; Héctor Gallardo-Rincón,
| | - Diana Gómez-Martín
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- *Correspondence: Diana Gómez-Martín, ; Héctor Gallardo-Rincón,
| |
Collapse
|
29
|
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
|
30
|
Seeßle J, Naujokat C, Oberacker P, Peters WH, Waterboer T, Müller B, Merle U. Reply to Peluso et al. Clin Infect Dis 2022; 74:2084-2085. [PMID: 34617996 DOI: 10.1093/cid/ciab892] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Jessica Seeßle
- Department of Internal Medicine IV, University Hospital Heidelberg, Heidelberg, Germany
| | - Cord Naujokat
- SYNLAB MVZ Laborzentrum Ettlingen GmbH, Ettlingen, Germany
| | - Phil Oberacker
- SYNLAB MVZ Laborzentrum Ettlingen GmbH, Ettlingen, Germany
| | | | - Tim Waterboer
- Infections and Cancer Epidemiology, German Cancer Research Center (Deutsches Krebsforschungszentrum [DKFZ]), Heidelberg, Germanyand
| | - Barbara Müller
- Department of Infectious Diseases, Virology, University Hospital Heidelberg, Heidelberg, Germany
| | - Uta Merle
- Department of Internal Medicine IV, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
31
|
Farzi R, Aghbash PS, Eslami N, Azadi A, Shamekh A, Hemmat N, Entezari-Maleki T, Baghi HB. The role of antigen-presenting cells in the pathogenesis of COVID-19. Pathol Res Pract 2022; 233:153848. [PMID: 35338971 PMCID: PMC8941975 DOI: 10.1016/j.prp.2022.153848] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/02/2022] [Accepted: 03/17/2022] [Indexed: 02/06/2023]
Abstract
Coronavirus Disease 2019 (COVID-19) is one of the three lethal coronavirus outbreaks in the recent two decades and a serious threat to global health all over the world. The principal feature of the COVID-19 infection is the so-called "cytokine storm" exaggerated molecular response to virus distribution, which plays massive tissue and organ injury roles. Immunological treatments, including monoclonal antibodies and vaccines, have been suggested as the main approaches in treating and preventing this disease. Therefore, a proper investigation of the roles of antigen-presenting cells (APCs) in the aforementioned immunological responses appears essential. The present review will provide detailed information about APCs' role in the infection and pathogenesis of SARS-CoV-2 and the effect of monoclonal antibodies in diagnosis and treatment.
Collapse
Affiliation(s)
- Rana Farzi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parisa Shiri Aghbash
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Narges Eslami
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Drug Applied Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arezou Azadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Shamekh
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nima Hemmat
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Drug Applied Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Taher Entezari-Maleki
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Bannazadeh Baghi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
32
|
Ulndreaj A, Wang M, Misaghian S, Paone L, Sigal GB, Stengelin M, Campbell C, Van Nynatten LR, Soosaipillai A, Ghorbani A, Mathew A, Fraser DD, Diamandis EP, Prassas I. Patients with severe COVID-19 do not have elevated autoantibodies against common diagnostic autoantigens. Clin Chem Lab Med 2022; 60:1116-1123. [PMID: 35475723 DOI: 10.1515/cclm-2022-0239] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/14/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Infection by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the causative pathogen of coronavirus disease 2019 (COVID-19) presents occasionally with an aberrant autoinflammatory response, including the presence of elevated circulating autoantibodies in some individuals. Whether the development of autoantibodies against self-antigens affects COVID-19 outcomes remains unclear. To better understand the prognostic role of autoantibodies in COVID-19, we quantified autoantibodies against 23 markers that are used for diagnosis of autoimmune disease. To this end, we used serum samples from patients with severe [intensive care unit (ICU)] and moderate (ward) COVID-19, across two to six consecutive time points, and compared autoantibody levels to uninfected healthy and ICU controls. METHODS Acute and post-acute serum (from 1 to 26 ICU days) was collected from 18 ICU COVID-19-positive patients at three to six time points; 18 ICU COVID-19-negative patients (sampled on ICU day 1 and 3); 21 ward COVID-19-positive patients (sampled on hospital day 1 and 3); and from 59 healthy uninfected controls deriving from two cohorts. Levels of IgG autoantibodies against 23 autoantigens, commonly used for autoimmune disease diagnosis, were measured in serum samples using MSD® U-PLEX electrochemiluminescence technology (MSD division Meso Scale Discovery®), and results were compared between groups. RESULTS There were no significant elevations of autoantibodies for any of the markers tested in patients with severe COVID-19. CONCLUSIONS Sample collections at longer time points should be considered in future studies, for assessing the possible development of autoantibody responses following infection with SARS-CoV-2.
Collapse
Affiliation(s)
- Antigona Ulndreaj
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Mingyue Wang
- Meso Scale Diagnostics, LLC. (MSD), Rockville, MD, USA
| | | | - Louis Paone
- Meso Scale Diagnostics, LLC. (MSD), Rockville, MD, USA
| | | | | | | | - Logan R Van Nynatten
- Lawson Health Research Institute, London, ON, Canada.,Department of Pediatrics, Clinical Neurological Sciences and Physiology and Pharmacology, Western University, London, ON, Canada
| | - Antoninus Soosaipillai
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON, Canada
| | - Atefeh Ghorbani
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Anu Mathew
- Meso Scale Diagnostics, LLC. (MSD), Rockville, MD, USA
| | - Douglas D Fraser
- Lawson Health Research Institute, London, ON, Canada.,Department of Pediatrics, Clinical Neurological Sciences and Physiology and Pharmacology, Western University, London, ON, Canada
| | - Eleftherios P Diamandis
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.,Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Department of Clinical Biochemistry, University Health Network, Toronto, ON, Canada
| | - Ioannis Prassas
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.,Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON, Canada
| |
Collapse
|
33
|
Wang JY, Zhang W, Roehrl VB, Roehrl MW, Roehrl MH. An Autoantigen Atlas From Human Lung HFL1 Cells Offers Clues to Neurological and Diverse Autoimmune Manifestations of COVID-19. Front Immunol 2022; 13:831849. [PMID: 35401574 PMCID: PMC8987778 DOI: 10.3389/fimmu.2022.831849] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 02/21/2022] [Indexed: 12/27/2022] Open
Abstract
COVID-19 is accompanied by a myriad of both transient and long-lasting autoimmune responses. Dermatan sulfate (DS), a glycosaminoglycan crucial for wound healing, has unique affinity for autoantigens (autoAgs) from apoptotic cells. DS-autoAg complexes are capable of stimulating autoreactive B cells and autoantibody production. We used DS-affinity proteomics to define the autoantigen-ome of lung fibroblasts and bioinformatics analyses to study the relationship between autoantigenic proteins and COVID-induced alterations. Using DS-affinity, we identified an autoantigen-ome of 408 proteins from human HFL1 cells, at least 231 of which are known autoAgs. Comparing with available COVID data, 352 proteins of the autoantigen-ome have thus far been found to be altered at protein or RNA levels in SARS-CoV-2 infection, 210 of which are known autoAgs. The COVID-altered proteins are significantly associated with RNA metabolism, translation, vesicles and vesicle transport, cell death, supramolecular fibrils, cytoskeleton, extracellular matrix, and interleukin signaling. They offer clues to neurological problems, fibrosis, smooth muscle dysfunction, and thrombosis. In particular, 150 altered proteins are related to the nervous system, including axon, myelin sheath, neuron projection, neuronal cell body, and olfactory bulb. An association with the melanosome is also identified. The findings from our study illustrate a connection between COVID infection and autoimmunity. The vast number of COVID-altered proteins with high intrinsic propensity to become autoAgs offers an explanation for the diverse autoimmune complications in COVID patients. The variety of autoAgs related to mRNA metabolism, translation, and vesicles suggests a need for long-term monitoring of autoimmunity in COVID. The COVID autoantigen atlas we are establishing provides a detailed molecular map for further investigation of autoimmune sequelae of the pandemic, such as "long COVID" syndrome. Summary Sentence An autoantigen-ome by dermatan sulfate affinity from human lung HFL1 cells may explain neurological and autoimmune manifestations of COVID-19.
Collapse
Affiliation(s)
| | - Wei Zhang
- Department of Gastroenterology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | | | | | - Michael H. Roehrl
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
34
|
Sapkota HR, Nune A. Long COVID from rheumatology perspective - a narrative review. Clin Rheumatol 2022; 41:337-348. [PMID: 34845562 PMCID: PMC8629735 DOI: 10.1007/s10067-021-06001-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/07/2021] [Accepted: 11/21/2021] [Indexed: 02/08/2023]
Abstract
Long-term sequel of acute COVID-19, commonly referred to as long COVID, has affected millions of patients worldwide. Long COVID patients display persistent or relapsing and remitting symptoms that include fatigue, breathlessness, cough, myalgia, arthralgia, sleep disturbance, cognitive impairment and skin rashes. Due to the shared clinical features, laboratory and imaging findings, long COVID could mimic rheumatic disease posing a diagnostic challenge. Our comprehensive literature review will help rheumatologist to be aware of long COVID manifestations and differentiating features from rheumatic diseases to ensure a timely and correct diagnosis is reached.
Collapse
Affiliation(s)
- Hem Raj Sapkota
- The Royal Wolverhampton Hospital NHS Trust, Wolverhampton, WV10 0QP UK
| | - Arvind Nune
- Southport and Ormskirk NHS Trust, Southport, PR8 6PN UK
| |
Collapse
|
35
|
Larionova R, Byvaltsev K, Kravtsova О, Takha E, Petrov S, Kazarian G, Valeeva A, Shuralev E, Mukminov M, Renaudineau Y, Arleevskaya M. SARS-Cov2 acute and post-active infection in the context of autoimmune and chronic inflammatory diseases. J Transl Autoimmun 2022; 5:100154. [PMID: 35434592 PMCID: PMC9005220 DOI: 10.1016/j.jtauto.2022.100154] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 03/31/2022] [Indexed: 12/11/2022] Open
Abstract
The clinical and immunological spectrum of acute and post-active COVID-19 syndrome overlaps with criteria used to characterize autoimmune diseases such as rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE). Indeed, following SARS-Cov2 infection, the innate immune response is altered with an initial delayed production of interferon type I (IFN-I), while the NF-kappa B and inflammasome pathways are activated. In lung and digestive tissues, an alternative and extrafollicular immune response against SARS-Cov2 takes place with, consequently, an altered humoral and memory T cell response leading to breakdown of tolerance with the emergence of autoantibodies. However, the risk of developing severe COVID-19 among SLE and RA patients did not exceed the general population except in those having pre-existing neutralizing autoantibodies against IFN-I. Treatment discontinuation rather than COVID-19 infection or vaccination increases the risk of developing flares. Last but not least, a limited number of case reports of individuals having developed SLE or RA following COVID-19 infection/vaccination have been reported. Altogether, the SARS-Cov2 pandemic represents an unique opportunity to investigate the dangerous interplay between the immune response against infectious agents and autoimmunity, and to better understand the triggering role of infection as a risk factor in autoimmune and chronic inflammatory disease development.
Collapse
Key Words
- ACE2, angiotensin converting enzyme 2
- ACPA, anti-cyclic citrullinated peptide autoAb
- ANA, antinuclear autoAb
- AutoAb, autoantibodies
- BAFF/BlySS, B-cell-activating factor/B lymphocyte stimulator
- CCL, chemokine ligand
- COVID-19, coronavirus disease 2019
- DMARDs, disease-modifying anti-rheumatic drugs
- E, envelope
- HEp-2, human epithelioma cell line 2
- IFN-I, interferon type I
- IFNAR, IFN-alpha receptors
- IL, interleukin
- IRF, interferon regulatory factor
- ISGs, IFN-stimulated genes
- ITP, immune-thrombocytopenic purpura
- Ig, immunoglobulin
- Infection
- Inflammation
- Jak, Janus kinase
- LDH, lactate dehydrogenase
- M, membrane
- MDA-5, melanoma differentiation-associated protein
- MERS-Cov, Middle East respiratory syndrome coronavirus
- MIS-C, multisystem inflammatory syndrome in children
- N, nucleocapsid
- NET, nuclear extracellular traps
- NF-κB, nuclear factor-kappa B
- NK, natural killer
- NLRP3, NOD-like receptor family
- Rheumatoid arthritis
- Risk factors
- SARS-Cov2
- Systemic lupus erythematosus
- T cell receptor, TLR
- Toll-like receptor, TMPRSS2
- aPL, antiphospholipid
- mAb, monoclonal Ab
- open reading frame, PACS
- pathogen-associated molecular patterns, pDC
- pattern recognition receptors, RA
- peptidylarginine deiminase 4, PAMPs
- plasmacytoid dendritic cells, PMN
- polymorphonuclear leukocytes, PRRs
- post-active COVID-19 syndrome, PAD-4
- primary Sjögren's syndrome, SLE
- pyrin domain containing 3, ORF
- reactive oxygen species, rt-PCR
- receptor binding domain, RF
- regulatory T cells, VDJ
- retinoic acid-inducible gene I, ROS
- reverse transcription polymerase chain reaction, S
- rheumatoid arthritis, RBD
- rheumatoid factor, RIG-I
- severe acute respiratory coronavirus 2, SjS
- signal transducer and activator of transcription, TCR
- single-stranded ribonucleic acid, STAT
- spike, SAD
- systemic autoimmune disease, SARS-Cov2
- systemic lupus erythematosus, SSc
- systemic sclerosis, ssRNA
- transmembrane serine protease 2, TNF
- tumor necrosis factor, Treg
- variable, diversity and joining Ig genes
Collapse
Affiliation(s)
- Regina Larionova
- Central Research Laboratory, Kazan State Medical Academy, Kazan, Russia
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia
| | - K Byvaltsev
- Institute of Fundamental Medicine, Kazan (Volga Region) Federal University, Kazan, Russia
| | - Оlga Kravtsova
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia
| | - Elena Takha
- Central Research Laboratory, Kazan State Medical Academy, Kazan, Russia
| | - Sergei Petrov
- Central Research Laboratory, Kazan State Medical Academy, Kazan, Russia
- Institute of Environmental Sciences, Kazan (Volga Region) Federal University, Kazan, Russia
| | - Gevorg Kazarian
- Central Research Laboratory, Kazan State Medical Academy, Kazan, Russia
| | - Anna Valeeva
- Central Research Laboratory, Kazan State Medical Academy, Kazan, Russia
| | - Eduard Shuralev
- Central Research Laboratory, Kazan State Medical Academy, Kazan, Russia
- Institute of Environmental Sciences, Kazan (Volga Region) Federal University, Kazan, Russia
- Kazan State Academy of Veterinary Medicine Named After N.E. Bauman, Kazan, Russia
| | - Malik Mukminov
- Central Research Laboratory, Kazan State Medical Academy, Kazan, Russia
- Institute of Environmental Sciences, Kazan (Volga Region) Federal University, Kazan, Russia
| | - Yves Renaudineau
- Central Research Laboratory, Kazan State Medical Academy, Kazan, Russia
- Laboratory of Immunology, CHU Purpan Toulouse, INSERM U1291, CNRS U5051, University Toulouse III, Toulouse, France
| | - Marina Arleevskaya
- Central Research Laboratory, Kazan State Medical Academy, Kazan, Russia
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia
| |
Collapse
|
36
|
Liu Y, Ebinger JE, Mostafa R, Budde P, Gajewski J, Walker B, Joung S, Wu M, Bräutigam M, Hesping F, Rupieper E, Schubert AS, Zucht HD, Braun J, Melmed GY, Sobhani K, Arditi M, Van Eyk JE, Cheng S, Fert-Bober J. Paradoxical sex-specific patterns of autoantibody response to SARS-CoV-2 infection. J Transl Med 2021; 19:524. [PMID: 34965855 PMCID: PMC8716184 DOI: 10.1186/s12967-021-03184-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 12/04/2021] [Indexed: 11/16/2022] Open
Abstract
Background Pronounced sex differences in the susceptibility and response to SARS-CoV-2 infection remain poorly understood. Emerging evidence has highlighted the potential importance of autoimmune activation in modulating the acute response and recovery trajectories following SARS-CoV-2 exposure. Given that immune-inflammatory activity can be sex-biased in the setting of severe COVID-19 illness, the aim of the study was to examine sex-specific autoimmune reactivity to SARS-CoV-2 in the absence of extreme clinical disease. Methods In this study, we assessed autoantibody (AAB) reactivity to 91 autoantigens previously linked to a range of classic autoimmune diseases in a cohort of 177 participants (65% women, 35% men, mean age of 35) with confirmed evidence of prior SARS-CoV-2 infection based on presence of antibody to the nucleocapsid protein of SARS-CoV-2. Data were compared to 53 pre-pandemic healthy controls (49% women, 51% men). For each participant, socio-demographic data, serological analyses, SARS-CoV-2 infection status and COVID-19 related symptoms were collected by an electronic survey of questions. The symptoms burden score was constructed based on the total number of reported symptoms (N = 21) experienced within 6 months prior to the blood draw, wherein a greater number of symptoms corresponded to a higher score and assigned as more severe burden. Results In multivariable analyses, we observed sex-specific patterns of autoreactivity associated with the presence or absence (as well as timing and clustering of symptoms) associated with prior COVID-19 illness. Whereas the overall AAB response was more prominent in women following asymptomatic infection, the breadth and extent of AAB reactivity was more prominent in men following at least mildly symptomatic infection. Notably, the observed reactivity included distinct antigens with molecular homology with SARS-CoV-2. Conclusion Our results reveal that prior SARS-CoV-2 infection, even in the absence of severe clinical disease, can lead to a broad AAB response that exhibits sex-specific patterns of prevalence and antigen selectivity. Further understanding of the nature of triggered AAB activation among men and women exposed to SARS-CoV-2 will be essential for developing effective interventions against immune-mediated sequelae of COVID-19. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03184-8.
Collapse
|
37
|
Lamprinos N, Ladomenou F, Stefanaki S, Foukarakis E, Vlachaki G. Pericarditis Following Recovery From COVID-19 Infection in a 15-Year-Old Boy: A Postinflammatory Immune-Mediated Presentation or a New-Onset Autoimmune Disease. Cureus 2021; 13:e19255. [PMID: 34881119 PMCID: PMC8643140 DOI: 10.7759/cureus.19255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2021] [Indexed: 11/22/2022] Open
Abstract
As the COVID-19 pandemic evolves, the medical community continues to report a variety of clinical manifestations of SARS-CoV-2 in the pediatric population. Although younger age groups experience less severe disease, attention is given to the immunologic manifestations of the disease. Pericarditis is a rare cardiac complication of COVID-19 infection. We discuss the first case of delayed presentation of pericarditis following recovery from COVID-19 infection in the pediatric population. A 15-year-old male adolescent presented to the emergency department (ED) with a two-day history of left-sided, sub-sternal chest pain that worsened during inspiration and a low-grade fever. Twenty days prior to this presentation, the patient experienced fever and was tested positive for SARS-CoV-2. His family history was remarkable for Hashimoto thyroiditis and rheumatoid arthritis, with his mother having experienced 18 episodes of pericarditis during the exacerbations of her disease. RT-PCR for SARS-CoV-2 was negative on this occasion and the serology assay identified positive IgG antibodies against the virus. The ECG was suggestive for pericarditis and the diagnosis was confirmed by the presence of pericardial effusion on ECHO. The rest of the aetiological investigations for pericarditis were negative. In view of the strong family history of autoimmunity, questions were raised in the medical team of our hospital regarding the etiology of his pericarditis and on whether it represented a postinflammatory immune-mediated presentation of SARS-CoV-2 or a new-onset autoimmune disease.
Collapse
Affiliation(s)
- Nikolas Lamprinos
- Department of Pediatrics, Venizeleion General Hospital, Heraklion Crete, GRC
| | - Fani Ladomenou
- Department of Pediatrics, Venizeleion General Hospital, Heraklion Crete, GRC
| | - Sofia Stefanaki
- Department of Pediatrics, Venizeleion General Hospital, Heraklion Crete, GRC
| | | | - Georgia Vlachaki
- Department of Pediatrics, Venizeleion General Hospital, Heraklion Crete, GRC
| |
Collapse
|
38
|
Peker BO, Şener AG, Kaptan Aydoğmuş F. Antinuclear antibodies (ANAs) detected by indirect immunofluorescence (IIF) method in acute COVID-19 infection; future roadmap for laboratory diagnosis. J Immunol Methods 2021; 499:113174. [PMID: 34737165 PMCID: PMC8556075 DOI: 10.1016/j.jim.2021.113174] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/06/2021] [Accepted: 10/20/2021] [Indexed: 01/04/2023]
Abstract
INTRODUCTION As in other viral infections, anti-nuclear antibodies (ANAs) are observed in SARS-CoV-2 infection. We investigated the presence of autoantibodies in acute COVID-19 and the association with early laboratory findings. MATERIALS AND METHODS We examined 50 sera (>18 years, 25 Female) from patients with acute COVID-19. ANAs (HEp-20-10 liver biochip), anti-neutrophil cytoplasmic antibody (ANCA, Europlus Granulocyte Mosaic 32) and anti-double stranded DNA were investigated with product of Euroimmune AG (Luebeck, Germany) by indirect immunofluorescence (IIF) method. Also, antibody against cyclic citrullinated peptide (anti-CCP) was examined by a chemiluminisens assay (Euroimmun AG, Luebeck, Germany). Samples from 50 blood bank donors collected before the COVID-19 pandemic were used as controls. RESULTS The IIF-ANA test was positive in 18% (N = 9/50) of the patients. The median time of sample collection was 7 days (range: 1-28 days) after diagnosis. ANA was positive in only one (2%) control sample. Five (55.5%) patients were ANA positive with a strong titer (3+). There was no relationship between antibody titration and time of sample collection (p = 0,55). Anti-CCP was detected in a nucleolar (3+) positive patient (2%). ANA was detected in 14.28% (N = 1/7, rods-rings (±), p = 0,78) of patients in the intensive care unit(ICU). Patients treated in the clinic have more and higher titers of ANA, mostly in nucleolar patterns, than ICU patients. CONCLUSIONS The variety of antibodies detected in acute COVID-19 and the uncertainty of how long they persist can lead to confusion, especially in the diagnosis of systemic autoimmune rheumatic diseases for IIF-ANA testing in immunology laboratories. Improvements in cell lines and methods will facilitate the diagnostic process.
Collapse
Affiliation(s)
- Bilal Olcay Peker
- Department of Medical Microbiology, İzmir Katip Çelebi University Atatürk Training And Research Hospital, İzmir, Turkey,Corresponding author at: Department of Medical Microbiology, İzmir Katip Çelebi University Atatürk Training and Research Hospital, Basın Sitesi, Karabağlar 35360, İzmir, Turkey
| | - Aslı Gamze Şener
- Department of Medical Microbiology, İzmir Katip Çelebi University Atatürk Training And Research Hospital, İzmir, Turkey
| | - Figen Kaptan Aydoğmuş
- Department of Infectious Diseases, İzmir Katip Çelebi University Atatürk Training And Research Hospital, İzmir, Turkey
| |
Collapse
|
39
|
Wintler T, Zherebtsov M, Carmack S, Muntean R, Hill SJ. Acute PR3-ANCA vasculitis in an asymptomatic COVID-19 teenager. JOURNAL OF PEDIATRIC SURGERY CASE REPORTS 2021; 75:102103. [PMID: 34729348 PMCID: PMC8554009 DOI: 10.1016/j.epsc.2021.102103] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 09/10/2021] [Indexed: 12/24/2022] Open
Abstract
We present the case of an acute onset ANCA positive vasculitis in an asymptomatic COVID-19 infected teenager, resulting in significant colonic damage. The patient was initially diagnosed with Henoch-Schönlein purpura and presented with worsening symptoms with significant necrosis of her perineum and rectum requiring surgical debridement and diverting colostomy. As a part of her work-up, she tested positive for COVID-19 total IgG/IgM antibodies and ANCA antibodies. This case complements previously reported cases of COVID-19 induced autoimmune disease in children but is novel in describing extensive intestinal disease as a result of an autoimmune vasculitis in a child.
Collapse
Key Words
- ANA, anti-nuclear antibody
- ANCA
- ANCA, antineutrophil cytoplasmic antibody
- CMV, Cytomegalovirus
- COVID-19
- COVID-19, coronavirus disease 2019
- EBV, Epstein-Barr virus
- HSP, Henoch Schönlein Purpura
- Henoch Schonlein purpura
- IgG, immunoglobulin G
- MIS-C, multisystem inflammatory syndrome in children
- NSAIDs, Non-steroidal anti-inflammatory drugs
- Vasculitis
- anti-MPO, myeloperoxidase antibody
- anti-PR3, proteinase 3 antibody
Collapse
Affiliation(s)
- Taylor Wintler
- Washington State University, Elson S. Floyd College of Medicine, Spokane, WA, USA
| | - Monica Zherebtsov
- Pediatric Gastroenterology Providence Digestive Health Institute, Spokane, WA, USA
| | | | - Rebecca Muntean
- Providence Rheumatology Providence Health and Services, Spokane, WA, USA
| | - Sarah J Hill
- Providence Sacred Heart Medical Center Pediatric Surgery, Spokane, WA, USA
| |
Collapse
|
40
|
Muratori P, Lenzi M, Muratori L, Granito A. Antinuclear antibodies in COVID 19. Clin Transl Sci 2021; 14:1627-1628. [PMID: 33932091 PMCID: PMC8239781 DOI: 10.1111/cts.13026] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/14/2022] Open
Affiliation(s)
- Paolo Muratori
- Division of Internal MedicineIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
- Department for the Science of the Quality of Life (QUVI)Alma Mater StudiorumUniversity of BolognaBolognaItaly
| | - Marco Lenzi
- Division of Internal MedicineIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
- Department of Medical and Surgical Sciences (DIMEC)Alma Mater StudiorumUniversity of BolognaBolognaItaly
| | - Luigi Muratori
- Division of Internal MedicineIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
- Department of Medical and Surgical Sciences (DIMEC)Alma Mater StudiorumUniversity of BolognaBolognaItaly
| | - Alessandro Granito
- Division of Internal MedicineIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
- Department of Medical and Surgical Sciences (DIMEC)Alma Mater StudiorumUniversity of BolognaBolognaItaly
| |
Collapse
|
41
|
Karami Fath M, Jahangiri A, Ganji M, Sefid F, Payandeh Z, Hashemi ZS, Pourzardosht N, Hessami A, Mard-Soltani M, Zakeri A, Rahbar MR, Khalili S. SARS-CoV-2 Proteome Harbors Peptides Which Are Able to Trigger Autoimmunity Responses: Implications for Infection, Vaccination, and Population Coverage. Front Immunol 2021; 12:705772. [PMID: 34447375 PMCID: PMC8383889 DOI: 10.3389/fimmu.2021.705772] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/23/2021] [Indexed: 12/16/2022] Open
Abstract
Autoimmune diseases (ADs) could occur due to infectious diseases and vaccination programs. Since millions of people are expected to be infected with SARS-CoV-2 and vaccinated against it, autoimmune consequences seem inevitable. Therefore, we have investigated the whole proteome of the SARS-CoV-2 for its ability to trigger ADs. In this regard, the entire proteome of the SARS-CoV-2 was chopped into more than 48000 peptides. The produced peptides were searched against the entire human proteome to find shared peptides with similar experimentally confirmed T-cell and B-cell epitopes. The obtained peptides were checked for their ability to bind to HLA molecules. The possible population coverage was calculated for the most potent peptides. The obtained results indicated that the SARS-CoV-2 and human proteomes share 23 peptides originated from ORF1ab polyprotein, nonstructural protein NS7a, Surface glycoprotein, and Envelope protein of SARS-CoV-2. Among these peptides, 21 peptides had experimentally confirmed equivalent epitopes. Amongst, only nine peptides were predicted to bind to HLAs with known global allele frequency data, and three peptides were able to bind to experimentally confirmed HLAs of equivalent epitopes. Given the HLAs which have already been reported to be associated with ADs, the ESGLKTIL, RYPANSIV, NVAITRAK, and RRARSVAS were determined to be the most harmful peptides of the SARS-CoV-2 proteome. It would be expected that the COVID-19 pandemic and the vaccination against this pathogen could significantly increase the ADs incidences, especially in populations harboring HLA-B*08:01, HLA-A*024:02, HLA-A*11:01 and HLA-B*27:05. The Southeast Asia, East Asia, and Oceania are at higher risk of AD development.
Collapse
Affiliation(s)
- Mohsen Karami Fath
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Abolfazl Jahangiri
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mahmoud Ganji
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Sefid
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Zahra Payandeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Sadat Hashemi
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Navid Pourzardosht
- Biochemistry Department, Guilan University of Medical Sciences, Rasht, Iran
| | - Anahita Hessami
- School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maysam Mard-Soltani
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Dezful University of Medical Sciences, Dezful, Iran
| | - Alireza Zakeri
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| | - Mohammad Reza Rahbar
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| |
Collapse
|
42
|
Foret T, Dufrost V, Salomon Du Mont L, Costa P, Lefevre B, Lacolley P, Regnault V, Zuily S, Wahl D. Systematic Review of Antiphospholipid Antibodies in COVID-19 Patients: Culprits or Bystanders? Curr Rheumatol Rep 2021; 23:65. [PMID: 34218350 PMCID: PMC8254447 DOI: 10.1007/s11926-021-01029-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2021] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW COVID-19 patients have a procoagulant state with a high prevalence of thrombotic events. The hypothesis of an involvement of antiphospholipid antibodies (aPL) has been suggested by several reports. Here, we reviewed 48 studies investigating aPL in COVID-19 patients. RECENT FINDINGS Prevalence of Lupus Anticoagulant (LA) ranged from 35% to 92% in ICU patients. Anti-cardiolipin (aCL) IgG and IgM were found in up to 52% and up to 40% of patients respectively. Anti-β2-glycoprotein I (aβ2-GPI) IgG and IgM were found in up to 39% and up to 34% of patients respectively. Between 1% and 12% of patients had a triple positive aPL profile. There was a high prevalence of aβ2-GPI and aCL IgA isotype. Two cohort studies found few persistent LA but more persistent solid phase assay aPL over time. aPL determination and their potential role is a real challenge for the treatment of this disease.
Collapse
Affiliation(s)
- Thomas Foret
- Université de Lorraine, INSERM, DCAC, F-54000, Nancy, France
| | - Virginie Dufrost
- Université de Lorraine, INSERM, DCAC, F-54000, Nancy, France
- CHRU-Nancy, Vascular Medicine Division and Regional Competence Center for Rare Vascular and Systemic Autoimmune Diseases, F-54000, Nancy, France
| | - Lucie Salomon Du Mont
- CHRU-Besancon, Vascular and Endovascular Surgery Department, F-25000, Besancon, France
- Université de Bourgogne Franche-Comté, EA3920, F-25000, Besancon, France
| | - Patricia Costa
- CHRU-Besancon, Vascular Medicine Unit, Vascular and Endovascular Surgery Department, F-25000, Besancon, France
| | - Benjamin Lefevre
- Université de Lorraine, CHRU-Nancy, Infectious and Tropical Diseases, F-54000, Nancy, France
- Université de Lorraine, APEMAC, F-54000, Nancy, France
| | | | | | - Stephane Zuily
- Université de Lorraine, INSERM, DCAC, F-54000, Nancy, France
- CHRU-Nancy, Vascular Medicine Division and Regional Competence Center for Rare Vascular and Systemic Autoimmune Diseases, F-54000, Nancy, France
| | - Denis Wahl
- Université de Lorraine, INSERM, DCAC, F-54000, Nancy, France.
- CHRU-Nancy, Vascular Medicine Division and Regional Competence Center for Rare Vascular and Systemic Autoimmune Diseases, F-54000, Nancy, France.
- INSERM UMR_S 1116 DCAC and CHRU-Nancy, Vascular Medicine Division and Regional Competence Center for Rare Auto-Immune Vascular Diseases, University of Lorraine, INSERM, University Hospital (CHRU) of Nancy, Nancy, France.
| |
Collapse
|
43
|
Ahmed S, Zimba O, Gasparyan AY. COVID-19 and the clinical course of rheumatic manifestations. Clin Rheumatol 2021; 40:2611-2619. [PMID: 33733315 PMCID: PMC7968918 DOI: 10.1007/s10067-021-05691-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/05/2021] [Accepted: 03/10/2021] [Indexed: 02/06/2023]
Abstract
The manifestations of COVID-19 have been evolving over time. Various post-COVID-19 syndromes are being recognised. Various viruses have been implicated in the pathogenesis of autoimmune diseases, and we expect a similar outcome with the severe acute respiratory syndrome-associated coronavirus-2 (SARS-CoV-2). The SARS-CoV-2 virus penetrates various tissues and organs and has a predisposition to lead to endotheliitis that may cause vascular manifestations including thrombosis. SARS-CoV-2 has been shown to activate Toll-like receptors and the complement system. It perpetuates NETosis and leads to autoantibody formation. These predispose to systemic autoimmunity. Both reactive arthritis and connective tissue disorders such as lupus and inflammatory myositis have been reported after COVID-19. Other reported autoimmune disorders include haemolytic anaemia, immune thrombocytopenia, cutaneous vasculitis, and Guillain Barré-like acute demyelinating disorders. The multi-system inflammatory syndrome in children and its adult counterpart are another post-COVID-19 entity that presents as an admixture of Kawasaki disease and staphylococcal toxic shock syndrome. Patients with preexisting rheumatic diseases may flare during the SARS-CoV-2 infection. They may develop novel autoimmune features also. The immune-suppressants used during the acute COVID-19 illness may confound the outcomes whereas comorbidities present in patients with rheumatic diseases may mask them. There is an urgent need to follow-up patients recovering from COVID and monitor autoantibody production in the context of rheumatic manifestations. Key Points • COVID-19 is associated with both innate and acquired immune reactions and production of various autoantibodies. • Various immune-mediated manifestations such as arthritis, myositis, haemolytic anaemia, thrombocytopenia, and acute demyelination may develop after COVID-19. • Longitudinal cohort data are warranted to describe, predict, and test prevent various rheumatic manifestations in post-COVID-19 subjects.
Collapse
Affiliation(s)
- Sakir Ahmed
- Department of Clinical Immunology & Rheumatology, Kalinga Institute of Medical Sciences (KIMS), KIIT University, Bhubaneswar, India
| | - Olena Zimba
- Department of Internal Medicine No. 2, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Armen Yuri Gasparyan
- Departments of Rheumatology and Research and Development, Russells Hall Hospital, Dudley, Dudley Group NHS Foundation Trust (Teaching Trust of the University of Birmingham, UK), Pensnett Road, Dudley, West Midlands DY1 2HQ UK
| |
Collapse
|
44
|
Favaloro EJ, Henry BM, Lippi G. COVID-19 and Antiphospholipid Antibodies: Time for a Reality Check? Semin Thromb Hemost 2021; 48:72-92. [PMID: 34130340 DOI: 10.1055/s-0041-1728832] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Antiphospholipid antibodies (aPL) comprise a panel of autoantibodies that reflect a potential prothrombotic risk in several autoimmune conditions, most notably antiphospholipid (antibody) syndrome (APS). aPL can be divided into those that form part of the laboratory criteria for APS, namely, lupus anticoagulant (LA), as well as anticardiolipin antibodies (aCL) and anti-β2-glycoprotein I antibodies (aβ2GPI) of the immunoglobulin G and M classes, and those that form a group considered as "noncriteria antibodies." The noncriteria antibodies include, for example, antiphosphatidylserine antibodies (aPS), antiprothrombin antibodies (aPT), and antiphosphatidylserine/prothrombin complex antibodies (aPS/PT). COVID-19 (coronavirus disease 2019) represents a prothrombotic disorder, and there have been several reports of various aPL being present in COVID-19 patients. There have also been similarities drawn between some of the pathophysiological features of COVID-19 and APS, in particular, the most severe form, catastrophic APS (CAPS). In this review, we critically appraise the literature on aPL and COVID-19. This is a companion piece to a separate review focused on LA. In the current review, we primarily concentrate on the so-called solid phase identifiable aPL, such as aCL and aβ2GPI, but also reflect on noncriteria aPL. We conclude that aPL positivity may be a feature of COVID-19, at least in some patients, but in general, identified "solid-phase" aPL are of low titer and not able to be well-linked to the thrombotic aspects of COVID-19. Also, most publications did not assess for aPL persistence, and where persistence was checked, the findings appeared to represent transient aPL. Importantly, high-titer aPL or multiple aPL positivity (including double, triple) were in the minority of COVID-19 presentations, and thus discount any widespread presence of APS, including the most severe form CAPS, in COVID-19 patients.
Collapse
Affiliation(s)
- Emmanuel J Favaloro
- Department of Haematology, Sydney Centres for Thrombosis and Haemostasis, Institute of Clinical Pathology and Medical Research (ICPMR), NSW Health Pathology, Westmead Hospital, Westmead, NSW, Australia.,School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW, Australia
| | - Brandon Michael Henry
- Cardiac Intensive Care Unit, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Giuseppe Lippi
- Section of Clinical Biochemistry, University of Verona, Verona, Italy
| |
Collapse
|
45
|
Aptamer BC 007's Affinity to Specific and Less-Specific Anti-SARS-CoV-2 Neutralizing Antibodies. Viruses 2021; 13:v13050932. [PMID: 34069827 PMCID: PMC8157297 DOI: 10.3390/v13050932] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/14/2021] [Accepted: 05/16/2021] [Indexed: 12/14/2022] Open
Abstract
COVID-19 is a pandemic respiratory disease that is caused by the highly infectious severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Anti-SARS-CoV-2 antibodies are essential weapons that a patient with COVID-19 has to combat the disease. When now repurposing a drug, namely an aptamer that interacts with SARS-CoV-2 proteins for COVID-19 treatment (BC 007), which is, however, a neutralizer of pathogenic autoantibodies in its original indication, the possibility of also binding and neutralizing anti-SARS-CoV-2 antibodies must be considered. Here, the highly specific virus-neutralizing antibodies have to be distinguished from the ones that also show cross-reactivity to tissues. The last-mentioned could be the origin of the widely reported SARS-CoV-2-induced autoimmunity, which should also become a target of therapy. We, therefore, used enzyme-linked immunosorbent assay (ELISA) technology to assess the binding of well-characterized publicly accessible anti-SARS-CoV-2 antibodies (CV07-209 and CV07-270) with BC 007. Nuclear magnetic resonance spectroscopy, isothermal calorimetric titration, and circular dichroism spectroscopy were additionally used to test the binding of BC 007 to DNA-binding sequence segments of these antibodies. BC 007 did not bind to the highly specific neutralizing anti-SARS-CoV-2 antibody but did bind to the less specific one. This, however, was a lot less compared to an autoantibody of its original indication (14.2%, range 11.0–21.5%). It was also interesting to see that the less-specific anti-SARS-CoV-2 antibody also showed a high background signal in the ELISA (binding on NeutrAvidin-coated or activated but noncoated plastic plate). These initial experiments suggest that the risk of binding and neutralizing highly specific anti-SARS CoV-2 antibodies by BC 007 should be low.
Collapse
|
46
|
Miedema J, Schreurs M, van der Sar-van der Brugge S, Paats M, Baart S, Bakker M, Hoek R, Dik WA, Endeman H, Van Der Velden V, van Gammeren A, Ermens A, Aerts JG, Thüsen JVD. Antibodies Against Angiotensin II Receptor Type 1 and Endothelin A Receptor Are Associated With an Unfavorable COVID19 Disease Course. Front Immunol 2021; 12:684142. [PMID: 34054876 PMCID: PMC8155664 DOI: 10.3389/fimmu.2021.684142] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/28/2021] [Indexed: 01/03/2023] Open
Abstract
Background Lung histopathology demonstrates vasculopathy in a subset of deceased COVID19 patients, which resembles histopathology observed in antibody-mediated lung transplant rejection. Autoantibodies against angiotensin II type 1 receptor (AT1R) and Endothelin receptor Type A (ETAR) have been demonstrated in antibody-mediated rejection and may also be associated with severe COVID19 infection. Objective To assess AT1R and ETAR auto-antibodies in COVID19 patients and controls, and explore their association with disease course. Methods 65 hospitalized patients with COVID19 infection were included. Clinical and laboratory findings were retrospectively assessed. Patients with unfavorable disease course, admitted at the intensive care unit and/or deceased during hospital admission (n=33) were compared to admitted COVID19 patients with favorable disease course (n=32). The presence of antinuclear antibodies (ANA) and auto-antibodies against AT1R or ETAR in peripheral blood were compared between COVID19 with unfavorable and favorable disease course and age matched controls (n=20). Results The presence of ANA was not significantly different between COVID19 patients with unfavorable (n=7/33; 21%) and favorable disease course (n=6/32; 19%) (p= 0.804) and controls (n=3/20; 15%). Auto-antibodies against AT1R were significantly increased in unfavorable disease course (median 14.59 U/mL, IQR 11.28 – 19.89) compared to favorable disease course (median 10.67 U/mL, IQR 8.55 – 13.0, p< 0.01). ETAR antibody titers were also significantly increased in unfavorable disease course (median 7.21, IQR 5.0 – 10.45) as compared to favorable disease course (median 4.0, IQR 3.0 – 6.0, p <0.05). Conclusion Auto-antibodies against AT1R and ETAR are significantly increased in COVID19 patients with an unfavorable disease course.
Collapse
Affiliation(s)
- Jelle Miedema
- Department of Pulmonology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Marco Schreurs
- Department of Immunology, Erasmus Medical Center, Rotterdam, Netherlands
| | | | - Marthe Paats
- Department of Pulmonology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Sara Baart
- Department of Biostatistics, Erasmus Medical Center, Rotterdam, Netherlands
| | - Marleen Bakker
- Department of Pulmonology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Rogier Hoek
- Department of Pulmonology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Willem Arnout Dik
- Department of Immunology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Henrik Endeman
- Department of Intensive Care Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | | | - Adriaan van Gammeren
- Department of Clinical Chemistry and Hematology, Amphia Hospital, Breda, Netherlands
| | - Antonius Ermens
- Department of Clinical Chemistry and Hematology, Amphia Hospital, Breda, Netherlands
| | - Joachim G Aerts
- Department of Pulmonology, Erasmus Medical Center, Rotterdam, Netherlands
| | | |
Collapse
|
47
|
Li M, Haque W, Vuppala S, Tobias E. Rare presentation of multisystem inflammatory syndrome in an adult associated with SARS-CoV-2 infection: unilateral neck swelling. BMJ Case Rep 2021; 14:14/5/e242392. [PMID: 33980565 PMCID: PMC8118037 DOI: 10.1136/bcr-2021-242392] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Multisystem inflammatory syndrome in adults (MIS-A) is a rare but often severe complication of SARS-CoV-2 infection. While several case reports about MIS-A in the setting of COVID-19 have been published since the term was first coined in June 2020, a clear description of the underlying pathophysiology and guideline-based recommendations on the diagnostic and therapeutic approach are lacking. What has been reported is that in the absence of severe respiratory illness, MIS-A can present with hypotension or shock, high-grade fever, abdominal pain, diarrhoea and severe weakness days to weeks after SARS-CoV-2 infection. Here, we present a case of a 28-year-old man who presented with a rarely described initial symptom: unilateral neck swelling. His presentation, disease progression and treatment course provide further information about MIS-A as a complication and in formulating diagnostic guidelines.
Collapse
Affiliation(s)
- Mozhu Li
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Waqas Haque
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Suchith Vuppala
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ethan Tobias
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
48
|
COVID-19-associated autoimmunity as a feature of acute respiratory failure. Intensive Care Med 2021; 47:801-804. [PMID: 33928414 PMCID: PMC8084710 DOI: 10.1007/s00134-021-06408-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 04/08/2021] [Indexed: 12/03/2022]
|
49
|
Wang JY, Zhang W, Roehrl MW, Roehrl VB, Roehrl MH. An autoantigen profile of human A549 lung cells reveals viral and host etiologic molecular attributes of autoimmunity in COVID-19. J Autoimmun 2021; 120:102644. [PMID: 33971585 PMCID: PMC8075847 DOI: 10.1016/j.jaut.2021.102644] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 04/08/2021] [Indexed: 12/13/2022]
Abstract
We aim to establish a comprehensive COVID-19 autoantigen atlas in order to understand autoimmune diseases caused by SARS-CoV-2 infection. Based on the unique affinity between dermatan sulfate and autoantigens, we identified 348 proteins from human lung A549 cells, of which 198 are known targets of autoantibodies. Comparison with current COVID data identified 291 proteins that are altered at protein or transcript level in SARS-CoV-2 infection, with 191 being known autoantigens. These known and putative autoantigens are significantly associated with viral replication and trafficking processes, including gene expression, ribonucleoprotein biogenesis, mRNA metabolism, translation, vesicle and vesicle-mediated transport, and apoptosis. They are also associated with cytoskeleton, platelet degranulation, IL-12 signaling, and smooth muscle contraction. Host proteins that interact with and that are perturbed by viral proteins are a major source of autoantigens. Orf3 induces the largest number of protein alterations, Orf9 affects the mitochondrial ribosome, and they and E, M, N, and Nsp proteins affect protein localization to membrane, immune responses, and apoptosis. Phosphorylation and ubiquitination alterations by viral infection define major molecular changes in autoantigen origination. This study provides a large list of autoantigens as well as new targets for future investigation, e.g., UBA1, UCHL1, USP7, CDK11A, PRKDC, PLD3, PSAT1, RAB1A, SLC2A1, platelet activating factor acetylhydrolase, and mitochondrial ribosomal proteins. This study illustrates how viral infection can modify host cellular proteins extensively, yield diverse autoantigens, and trigger a myriad of autoimmune sequelae. Our work provides a rich resource for studies into “long COVID” and related autoimmune sequelae.
Collapse
Affiliation(s)
| | - Wei Zhang
- Department of Gastroenterology, Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | | | | | - Michael H Roehrl
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, USA.
| |
Collapse
|
50
|
Wang JY, Zhang W, Roehrl VB, Roehrl MW, Roehrl MH. An Autoantigen-ome from HS-Sultan B-Lymphoblasts Offers a Molecular Map for Investigating Autoimmune Sequelae of COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.04.05.438500. [PMID: 33851168 PMCID: PMC8043459 DOI: 10.1101/2021.04.05.438500] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
To understand how COVID-19 may induce autoimmune diseases, we have been compiling an atlas of COVID-autoantigens (autoAgs). Using dermatan sulfate (DS) affinity enrichment of autoantigenic proteins extracted from HS-Sultan lymphoblasts, we identified 362 DS-affinity proteins, of which at least 201 (56%) are confirmed autoAgs. Comparison with available multi-omic COVID data shows that 315 (87%) of the 362 proteins are affected in SARS-CoV-2 infection via altered expression, interaction with viral components, or modification by phosphorylation or ubiquitination, at least 186 (59%) of which are known autoAgs. These proteins are associated with gene expression, mRNA processing, mRNA splicing, translation, protein folding, vesicles, and chromosome organization. Numerous nuclear autoAgs were identified, including both classical ANAs and ENAs of systemic autoimmune diseases and unique autoAgs involved in the DNA replication fork, mitotic cell cycle, or telomerase maintenance. We also identified many uncommon autoAgs involved in nucleic acid and peptide biosynthesis and nucleocytoplasmic transport, such as aminoacyl-tRNA synthetases. In addition, this study found autoAgs that potentially interact with multiple SARS-CoV-2 Nsp and Orf components, including CCT/TriC chaperonin, insulin degrading enzyme, platelet-activating factor acetylhydrolase, and the ezrin-moesin-radixin family. Furthermore, B-cell-specific IgM-associated ER complex (including MBZ1, BiP, heat shock proteins, and protein disulfide-isomerases) is enriched by DS-affinity and up-regulated in B-cells of COVID-19 patients, and a similar IgH-associated ER complex was also identified in autoreactive pre-B1 cells in our previous study, which suggests a role of autoreactive B1 cells in COVID-19 that merits further investigation. In summary, this study demonstrates that virally infected cells are characterized by alterations of proteins with propensity to become autoAgs, thereby providing a possible explanation for infection-induced autoimmunity. The COVID autoantigen-ome provides a valuable molecular resource and map for investigation of COVID-related autoimmune sequelae and considerations for vaccine design.
Collapse
Affiliation(s)
| | - Wei Zhang
- Department of Gastroenterology, Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | | | | | - Michael H. Roehrl
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, USA
| |
Collapse
|