1
|
Chatterjee S, Bhattacharya M, Saxena S, Lee SS, Chakraborty C. Autoantibodies in COVID-19 and Other Viral Diseases: Molecular, Cellular, and Clinical Perspectives. Rev Med Virol 2024; 34:e2583. [PMID: 39289528 DOI: 10.1002/rmv.2583] [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: 09/21/2023] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/19/2024]
Abstract
Autoantibodies are immune system-produced antibodies that wrongly target the body's cells and tissues for attack. The COVID-19 pandemic has made it possible to link autoantibodies to both the severity of pathogenic infection and the emergence of several autoimmune diseases after recovery from the infection. An overview of autoimmune disorders and the function of autoantibodies in COVID-19 and other infectious diseases are discussed in this review article. We also investigated the different categories of autoantibodies found in COVID-19 and other infectious diseases including the potential pathways by which they contribute to the severity of the illness. Additionally, it also highlights the probable connection between vaccine-induced autoantibodies and their adverse outcomes. The review also discusses the therapeutic perspectives of autoantibodies. This paper advances our knowledge about the intricate interaction between autoantibodies and COVID-19 by thoroughly assessing the most recent findings.
Collapse
Affiliation(s)
- Srijan Chatterjee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, South Korea
| | | | - Sanskriti Saxena
- Division of Biology, Indian Institute of Science Education and Research-Tirupati, Tirupati, India
| | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, South Korea
| | - Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, India
| |
Collapse
|
2
|
Bellucci M, Bozzano FM, Castellano C, Pesce G, Beronio A, Farshchi AH, Limongelli A, Uccelli A, Benedetti L, De Maria A. Post-SARS-CoV-2 infection and post-vaccine-related neurological complications share clinical features and the same positivity to anti-ACE2 antibodies. Front Immunol 2024; 15:1398028. [PMID: 39148725 PMCID: PMC11324485 DOI: 10.3389/fimmu.2024.1398028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 07/15/2024] [Indexed: 08/17/2024] Open
Abstract
Introduction A potential overlap in symptoms between post-acute COVID-19 syndrome and post-COVID-19 vaccination syndrome has been noted. We report a paired description of patients presenting with similar manifestations involving the central (CNS) or peripheral nervous system (PNS) following SARS-CoV-2 infection or vaccination, suggesting that both may have triggered similar immune-mediated neurological disorders in the presence of anti-idiotype antibodies directed against the ACE2 protein. Patients and methods Four patients exhibited overlapping neurological manifestations following SARS-CoV-2 infection or vaccination: radiculitis, Guillain-Barré syndrome, and MRI-negative myelitis, respectively, sharing positivity for anti-ACE2 antibodies. Autoantibodies against AQP-4, MOG, GlyR, GAD, and amphiphysin, onconeural antibodies for CNS syndromes, and anti-ganglioside antibodies for PNS syndromes tested negative in all patients. Discussion Anti-idiotype antibodies against ACE2 have been detected in patients who recovered from COVID-19 infection, and it has been hypothesized that such antibodies may mediate adverse events following SARS-CoV-2 infection or vaccination, resulting in the activation of the immune system against cells expressing ACE2, such as neurons. Our data reveal clinically overlapping syndromes triggered by SARS-CoV-2 infection or vaccination, sharing positivity for anti-ACE2 antibodies. Their presence, in the absence of other classic autoimmune markers of CNS or PNS involvement, suggests that they might play an active role in the context of an aberrant immune response. Conclusion Anti-idiotype antibodies directed against ACE2 may be triggered by both SARS-CoV-2 infection and vaccination, possibly contributing to neurological autoimmune manifestations. Their pathogenic role, however, remains to be demonstrated in large-scale, more structured studies.
Collapse
Affiliation(s)
- Margherita Bellucci
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genoa, Italy
| | - Federica Maria Bozzano
- Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Ospedale Policlinico San Martino, Genova, Italy
| | - Chiara Castellano
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genoa, Italy
| | - Giampaola Pesce
- Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Ospedale Policlinico San Martino, Genova, Italy
| | | | | | | | - Antonio Uccelli
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genoa, Italy
- Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Ospedale Policlinico San Martino, Genova, Italy
| | - Luana Benedetti
- Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Ospedale Policlinico San Martino, Genova, Italy
| | - Andrea De Maria
- Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Ospedale Policlinico San Martino, Genova, Italy
- Department of Health Sciences (DISSAL), University of Genova, Genoa, Italy
| |
Collapse
|
3
|
Tsay GJ, Zouali M. Cellular pathways and molecular events that shape autoantibody production in COVID-19. J Autoimmun 2024; 147:103276. [PMID: 38936147 DOI: 10.1016/j.jaut.2024.103276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/26/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
A hallmark of COVID-19 is the variety of complications that follow SARS-CoV-2 infection in some patients, and that target multiple organs and tissues. Also remarkable are the associations with several auto-inflammatory disorders and the presence of autoantibodies directed to a vast array of antigens. The processes underlying autoantibody production in COVID-19 have not been completed deciphered. Here, we review mechanisms involved in autoantibody production in COVID-19, multisystem inflammatory syndrome in children, and post-acute sequelae of COVID19. We critically discuss how genomic integrity, loss of B cell tolerance to self, superantigen effects of the virus, and extrafollicular B cell activation could underly autoantibody proaction in COVID-19. We also offer models that may account for the pathogenic roles of autoantibodies in the promotion of inflammatory cascades, thromboembolic phenomena, and endothelial and vascular deregulations.
Collapse
Affiliation(s)
- Gregory J Tsay
- Division of Immunology and Rheumatology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan; College of Medicine, China Medical University, Taichung, Taiwan
| | - Moncef Zouali
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.
| |
Collapse
|
4
|
Sanges S, Tian W, Dubucquoi S, Chang JL, Collet A, Launay D, Nicolls MR. B-cells in pulmonary arterial hypertension: friend, foe or bystander? Eur Respir J 2024; 63:2301949. [PMID: 38485150 PMCID: PMC11043614 DOI: 10.1183/13993003.01949-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 03/01/2024] [Indexed: 04/22/2024]
Abstract
There is an unmet need for new therapeutic strategies that target alternative pathways to improve the prognosis of patients with pulmonary arterial hypertension (PAH). As immunity has been involved in the development and progression of vascular lesions in PAH, we review the potential contribution of B-cells in its pathogenesis and evaluate the relevance of B-cell-targeted therapies. Circulating B-cell homeostasis is altered in PAH patients, with total B-cell lymphopenia, abnormal subset distribution (expansion of naïve and antibody-secreting cells, reduction of memory B-cells) and chronic activation. B-cells are recruited to the lungs through local chemokine secretion, and activated by several mechanisms: 1) interaction with lung vascular autoantigens through cognate B-cell receptors; 2) costimulatory signals provided by T follicular helper cells (interleukin (IL)-21), type 2 T helper cells and mast cells (IL-4, IL-6 and IL-13); and 3) increased survival signals provided by B-cell activating factor pathways. This activity results in the formation of germinal centres within perivascular tertiary lymphoid organs and in the local production of pathogenic autoantibodies that target the pulmonary vasculature and vascular stabilisation factors (including angiotensin-II/endothelin-1 receptors and bone morphogenetic protein receptors). B-cells also mediate their effects through enhanced production of pro-inflammatory cytokines, reduced anti-inflammatory properties by regulatory B-cells, immunoglobulin (Ig)G-induced complement activation, and IgE-induced mast cell activation. Precision-medicine approaches targeting B-cell immunity are a promising direction for select PAH conditions, as suggested by the efficacy of anti-CD20 therapy in experimental models and a trial of rituximab in systemic sclerosis-associated PAH.
Collapse
Affiliation(s)
- Sébastien Sanges
- Univ. Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
- INSERM, F-59000 Lille, France
- CHU Lille, Département de Médecine Interne et Immunologie Clinique, F-59000 Lille, France
- Centre National de Référence Maladies Auto-immunes Systémiques Rares du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), F-59000 Lille, France
- Health Care Provider of the European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases Network (ReCONNET), F-59000 Lille, France
- Veteran Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, School of Medicine, Stanford, CA, USA
- Both authors contributed equally and share co-first authorship
| | - Wen Tian
- Veteran Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, School of Medicine, Stanford, CA, USA
- Both authors contributed equally and share co-first authorship
| | - Sylvain Dubucquoi
- Univ. Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
- INSERM, F-59000 Lille, France
- CHU Lille, Institut d'Immunologie, Pôle de Biologie Pathologie Génétique, F-59000 Lille, France
| | - Jason L Chang
- Veteran Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, School of Medicine, Stanford, CA, USA
| | - Aurore Collet
- Univ. Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
- INSERM, F-59000 Lille, France
- CHU Lille, Institut d'Immunologie, Pôle de Biologie Pathologie Génétique, F-59000 Lille, France
| | - David Launay
- Univ. Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
- INSERM, F-59000 Lille, France
- CHU Lille, Département de Médecine Interne et Immunologie Clinique, F-59000 Lille, France
- Centre National de Référence Maladies Auto-immunes Systémiques Rares du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), F-59000 Lille, France
- Health Care Provider of the European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases Network (ReCONNET), F-59000 Lille, France
- Veteran Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, School of Medicine, Stanford, CA, USA
- Both authors contributed equally and share co-last authorship
| | - Mark R Nicolls
- Veteran Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, School of Medicine, Stanford, CA, USA
- Both authors contributed equally and share co-last authorship
| |
Collapse
|
5
|
Kieber-Emmons T. The Gift That Keeps on Giving. Monoclon Antib Immunodiagn Immunother 2024; 43:33-34. [PMID: 38593440 DOI: 10.1089/mab.2024.29018.editorial] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024] Open
|
6
|
Collins CP, Longo DL, Murphy WJ. The immunobiology of SARS-CoV-2 infection and vaccine responses: potential influences of cross-reactive memory responses and aging on efficacy and off-target effects. Front Immunol 2024; 15:1345499. [PMID: 38469293 PMCID: PMC10925677 DOI: 10.3389/fimmu.2024.1345499] [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: 11/27/2023] [Accepted: 02/12/2024] [Indexed: 03/13/2024] Open
Abstract
Immune responses to both SARS-CoV-2 infection and its associated vaccines have been highly variable within the general population. The increasing evidence of long-lasting symptoms after resolution of infection, called post-acute sequelae of COVID-19 (PASC) or "Long COVID," suggests that immune-mediated mechanisms are at play. Closely related endemic common human coronaviruses (hCoV) can induce pre-existing and potentially cross-reactive immunity, which can then affect primary SARS-CoV-2 infection, as well as vaccination responses. The influence of pre-existing immunity from these hCoVs, as well as responses generated from original CoV2 strains or vaccines on the development of new high-affinity responses to CoV2 antigenic viral variants, needs to be better understood given the need for continuous vaccine adaptation and application in the population. Due in part to thymic involution, normal aging is associated with reduced naïve T cell compartments and impaired primary antigen responsiveness, resulting in a reliance on the pre-existing cross-reactive memory cell pool which may be of lower affinity, restricted in diversity, or of shorter duration. These effects can also be mediated by the presence of down-regulatory anti-idiotype responses which also increase in aging. Given the tremendous heterogeneity of clinical data, utilization of preclinical models offers the greatest ability to assess immune responses under a controlled setting. These models should now involve prior antigen/viral exposure combined with incorporation of modifying factors such as age on immune responses and effects. This will also allow for mechanistic dissection and understanding of the different immune pathways involved in both SARS-CoV-2 pathogen and potential vaccine responses over time and how pre-existing memory responses, including potential anti-idiotype responses, can affect efficacy as well as potential off-target effects in different tissues as well as modeling PASC.
Collapse
Affiliation(s)
- Craig P. Collins
- Graduate Program in Immunology, University of California (UC) Davis, Davis, CA, United States
| | - Dan L. Longo
- Harvard Medical School, Brigham and Women’s Hospital, Boston, MA, United States
| | - William J. Murphy
- Departments of Dermatology and Internal Medicine (Hematology/Oncology), University of California (UC) Davis School of Medicine, Sacramento, CA, United States
| |
Collapse
|
7
|
Papavassiliou KA, Gogou VA, Papavassiliou AG. Angiotensin-Converting Enzyme 2 (ACE2) Signaling in Pulmonary Arterial Hypertension: Underpinning Mechanisms and Potential Targeting Strategies. Int J Mol Sci 2023; 24:17441. [PMID: 38139269 PMCID: PMC10744156 DOI: 10.3390/ijms242417441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a debilitating progressive disease characterized by excessive pulmonary vasoconstriction and abnormal vascular remodeling processes that lead to right-ventricular heart failure and, ultimately, death. Although our understanding of its pathophysiology has advanced and several treatment modalities are currently available for the management of PAH patients, none are curative and the prognosis remains poor. Therefore, further research is required to decipher the molecular mechanisms associated with PAH. Angiotensin-converting enzyme 2 (ACE2) plays an important role through its vasoprotective functions in cardiopulmonary homeostasis, and accumulating preclinical and clinical evidence shows that the upregulation of the ACE2/Angiotensin-(1-7)/MAS1 proto-oncogene, G protein-coupled receptor (Mas 1 receptor) signaling axis is implicated in the pathophysiology of PAH. Herein, we highlight the molecular mechanisms of ACE2 signaling in PAH and discuss its potential as a therapeutic target.
Collapse
Affiliation(s)
- Kostas A. Papavassiliou
- First University Department of Respiratory Medicine, ‘Sotiria’ Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Vassiliki A. Gogou
- First University Department of Respiratory Medicine, ‘Sotiria’ Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
8
|
Tsoi JYH, Cai J, Situ J, Lam WJ, Shun EHK, Leung JKY, Chen LL, Chan BPC, Yeung ML, Li X, Chan KH, Wong JSC, Kwan MYW, To KKW, Yuen KY, Sridhar S. Autoantibodies against angiotensin-converting enzyme 2 (ACE2) after COVID-19 infection or vaccination. J Med Virol 2023; 95:e29313. [PMID: 38100626 DOI: 10.1002/jmv.29313] [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: 10/10/2023] [Revised: 10/31/2023] [Accepted: 11/28/2023] [Indexed: 12/17/2023]
Abstract
Autoantibodies against angiotensin-converting enzyme 2 (ACE2) are frequently reported in patients during coronavirus disease 2019 (COVID-19) with evidence for a pathogenic role in severe infection. However, little is known of the prevalence or clinical significance of ACE2 autoantibodies in late convalescence or following COVID-19 vaccination. In this study, we measured ACE2 autoantibodies in a cohort of 182 COVID-19 convalescent patients, 186 COVID-19 vaccine recipients, and 43 adolescents with post-mRNA vaccine myopericarditis using two ACE2 enzymatic immunoassays (EIAs). ACE2 IgM autoantibody EIA median optical densities (ODs) were lower in convalescent patients than pre-COVID-19 control samples with only 2/182 (1.1%) convalescents testing positive. Similarly, only 3/182 (1.6%) convalescent patients tested positive for ACE2 IgG, but patients with history of moderate-severe COVID-19 tended to have significantly higher median ODs than controls and mild COVID-19 patients. In contrast, ACE2 IgG antibodies were detected in 10/186 (5.4%) COVID-19 vaccine recipients after two doses of vaccination. Median ACE2 IgG EIA ODs of vaccine recipients were higher than controls irrespective of the vaccine platform used (inactivated or mRNA). ACE2 IgG ODs were not correlated with surrogate neutralizing antibody levels in vaccine recipients. ACE2 IgG levels peaked at day 56 post-first dose and declined within 12 months to baseline levels in vaccine recipients. Presence of ACE2 antibodies was not associated with adverse events following immunization including myopericarditis. One convalescent patient with ACE2 IgG developed Guillain-Barre syndrome, but causality was not established. ACE2 autoantibodies are observed in COVID-19 vaccine recipients and convalescent patients, but are likely innocuous.
Collapse
Affiliation(s)
- James Yiu Hung Tsoi
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Jianpiao Cai
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Jianwen Situ
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Winston Jim Lam
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Estie Hon Kiu Shun
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Centre for Virology, Vaccinology and Therapeutics, The University of Hong Kong, Hong Kong, China
| | - Joy Ka Yi Leung
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Lin Lei Chen
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Brian Pui Chun Chan
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Man Lung Yeung
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Xin Li
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Kwok Hung Chan
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Joshua Sung Chih Wong
- Department of Paediatrics and Adolescent Medicine, Princess Margaret Hospital, Hong Kong, China
| | - Mike Yat Wah Kwan
- Department of Paediatrics and Adolescent Medicine, Princess Margaret Hospital, Hong Kong, China
| | - Kelvin Kai Wang To
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Centre for Virology, Vaccinology and Therapeutics, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
- Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kwok Yung Yuen
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Centre for Virology, Vaccinology and Therapeutics, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Hong Kong, China
- Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Siddharth Sridhar
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| |
Collapse
|
9
|
Zi-Yang Y, Nanshan X, Dongling L, Tao H, Yigao H, Danqing Y, Caojin Z. ACE2 gene polymorphisms are associated with elevated pulmonary artery pressure in congenital heart diseases. Gene 2023; 882:147642. [PMID: 37454747 DOI: 10.1016/j.gene.2023.147642] [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: 05/09/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUNDS Angiotensin converting enzyme 2 (ACE2) polymorphisms are related to the occurrence and prognosis of cardiovascular disease. However, whether ACE2 polymorphisms also affect pulmonary circulation in congenital heart disease (CHD) remains unclear. Thus, we investigated the relationship between ACE2 single nucleotide polymorphism (SNPs) and pulmonary circulation in CHD patients of Chinese Han ethnicity. METHODS Enrolled CHD patients (n = 367) underwent gene sequencing of ACE2 SNPs rs2074192, rs2285666, and rs2106809. Patients with pulmonary hypertension were further examined for detailed hemodynamics. RESULTS Female heterozygous patients had worse pulmonary circulation hemodynamic parameters compared to those of homozygotes. Female CHD patients with the CCA (OR = 0.53, 95% CI: 0.32-0.88) or CCG (OR = 0.59, 95% CI: 0.35-0.99) haplotype had a lower risk of elevated pulmonary artery pressure. CONCLUSION In female CHD patients, ACE2 SNPs are related to pulmonary circulation hemodynamics. Female CHD patients with the CCA and CCG haplotype had a lower risk of pulmonary hypertension.
Collapse
Affiliation(s)
- Yang Zi-Yang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Xie Nanshan
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, Guangdong, China
| | - Luo Dongling
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, Guangdong, China
| | - Huang Tao
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, Guangdong, China
| | - Huang Yigao
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, Guangdong, China
| | - Yu Danqing
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China.
| | - Zhang Caojin
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, Guangdong, China.
| |
Collapse
|
10
|
Lichtenstein B, Zheng Y, Gjertson D, Ferbas KG, Rimoin AW, Yang OO, Aldrovandi GM, Schaenman JM, Reed EF, Fulcher JA. Vascular and Non-HLA autoantibody profiles in hospitalized patients with COVID-19. Front Immunol 2023; 14:1197326. [PMID: 37398658 PMCID: PMC10309004 DOI: 10.3389/fimmu.2023.1197326] [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: 03/30/2023] [Accepted: 05/23/2023] [Indexed: 07/04/2023] Open
Abstract
Introduction Severe COVID-19 illness is characterized by an overwhelming immune hyperactivation. Autoantibodies against vascular, tissue, and cytokine antigens have been detected across the spectrum of COVID-19. How these autoantibodies correlate with COVID-19 severity is not fully defined. Methods We performed an exploratory study to investigate the expression of vascular and non-HLA autoantibodies in 110 hospitalized patients with COVID-19 ranging from moderate to critically ill. Relationships between autoantibodies and COVID- 19 severity and clinical risk factors were examined using logistic regression analysis. Results There were no absolute differences in levels of expression of autoantibodies against angiotensin II receptor type 1 (AT1R) or endothelial cell proteins between COVID-19 severity groups. AT1R autoantibody expression also did not differ by age, sex, or diabetes status. Using a multiplex panel of 60 non- HLA autoantigens we did identify seven autoantibodies that differed by COVID-19 severity including myosin (myosin; p=0.02), SHC-transforming protein 3 (shc3; p=0.07), peroxisome proliferator-activated receptor gamma coactivator 1-beta (perc; p=0.05), glial-cell derived neurotrophic factor (gdnf; p=0.07), enolase 1 (eno1; p=0.08), latrophilin-1 (lphn1; p=0.08), and collagen VI (coll6; p=0.05) with greater breadth and higher expression levels seen in less severe COVID-19. Discussion Overall, we found that patients hospitalized with COVID-19 demonstrate evidence of auto-reactive antibodies targeting endothelial cells, angiotensin II receptors, and numerous structural proteins including collagens. Phenotypic severity did not correlate with specific autoantibodies. This exploratory study underscores the importance of better understanding of the role of autoimmunity in COVID-19 disease and sequelae.
Collapse
Affiliation(s)
- Brian Lichtenstein
- Division of Hospital Medicine, Department of Internal Medicine, Sharp Rees-Stealy Medical Group, Sharp Healthcare, San Diego, CA, United States
| | - Ying Zheng
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - David Gjertson
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
- Department of Biostatistics, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, United States
| | - Kathie G. Ferbas
- Division of Infectious Diseases, Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Anne W. Rimoin
- Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, United States
| | - Otto O. Yang
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Grace M. Aldrovandi
- Division of Infectious Diseases, Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Joanna M. Schaenman
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Elaine F. Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Jennifer A. Fulcher
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
- Infectious Diseases Section, Veterans Affairs (VA) Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| |
Collapse
|
11
|
Abstract
The current epidemic of corona virus disease (COVID-19) has resulted in an immense health burden that became the third leading cause of death and potentially contributed to a decline in life expectancy in the United States. The severe acute respiratory syndrome-related coronavirus-2 binds to the surface-bound peptidase angiotensin-converting enzyme 2 (ACE2, EC 3.4.17.23) leading to tissue infection and viral replication. ACE2 is an important enzymatic component of the renin-angiotensin system (RAS) expressed in the lung and other organs. The peptidase regulates the levels of the peptide hormones Ang II and Ang-(1-7), which have distinct and opposing actions to one another, as well as other cardiovascular peptides. A potential consequence of severe acute respiratory syndrome-related coronavirus-2 infection is reduced ACE2 activity by internalization of the viral-ACE2 complex and subsequent activation of the RAS (higher ratio of Ang II:Ang-[1-7]) that may exacerbate the acute inflammatory events in COVID-19 patients and possibly contribute to the effects of long COVID-19. Moreover, COVID-19 patients present with an array of autoantibodies to various components of the RAS including the peptide Ang II, the enzyme ACE2, and the AT1 AT2 and Mas receptors. Greater disease severity is also evident in male COVID-19 patients, which may reflect underlying sex differences in the regulation of the 2 distinct functional arms of the RAS. The current review provides a critical evaluation of the evidence for an activated RAS in COVID-19 subjects and whether this system contributes to the greater severity of severe acute respiratory syndrome-related coronavirus-2 infection in males as compared with females.
Collapse
Affiliation(s)
- Mark C. Chappell
- Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC
| |
Collapse
|
12
|
Vilibic-Cavlek T, Bogdanic M, Borko E, Hruskar Z, Zilic D, Ferenc T, Tabain I, Barbic L, Vujica Ferenc M, Ferencak I, Stevanovic V. Detection of SARS-CoV-2 Antibodies: Comparison of Enzyme Immunoassay, Surrogate Neutralization and Virus Neutralization Test. Antibodies (Basel) 2023; 12:antib12020035. [PMID: 37218901 DOI: 10.3390/antib12020035] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 04/03/2023] [Accepted: 05/04/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND Since sensitivity and specificity vary widely between tests, SARS-CoV-2 serology results should be interpreted with caution. METHODS The study included serum samples from patients who had recovered from COVID-19 (n = 71), individuals vaccinated against SARS-CoV-2 (n = 84), and asymptomatic individuals (n = 33). All samples were tested for the presence of binding antibodies (enzyme immunoassay; EIA), neutralizing (NT) antibodies (virus neutralization test; VNT), and surrogate NT (sNT) antibodies (surrogate virus neutralization test; sVNT) of SARS-CoV-2. RESULTS SARS-CoV-2-binding antibodies were detected in 71 (100%) COVID-19 patients, 77 (91.6%) vaccinated individuals, and 4 (12.1%) control subjects. Among EIA-positive samples, VNT was positive (titer ≥ 8) in 100% of COVID-19 patients and 63 (75.0%) of the vaccinated individuals, while sVNT was positive (>30% inhibition) in 62 (87.3%) patients and 59 (70.2%) vaccinated individuals. The analysis of antibody levels showed a significant moderate positive correlation between EIA and VNT, a moderate positive correlation between EIA and sVNT, and a strong positive correlation between VNT and sVNT. The proportion of positive sVNT detection rate was associated with VNT titer. The lowest positivity (72.4%/70.8%) was detected in samples with low NT titers (8/16) and increased progressively from 88.2% in samples with titer 32 to 100% in samples with titer 256. CONCLUSIONS sVNT appeared to be a reliable method for the assessment COVID-19 serology in patients with high antibody levels, while false-negative results were frequently observed in patients with low NT titers.
Collapse
Affiliation(s)
- Tatjana Vilibic-Cavlek
- Department of Virology, Croatian Institute of Public Health, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Maja Bogdanic
- Department of Virology, Croatian Institute of Public Health, 10000 Zagreb, Croatia
| | - Ema Borko
- Department of Virology, Croatian Institute of Public Health, 10000 Zagreb, Croatia
| | - Zeljka Hruskar
- Department of Virology, Croatian Institute of Public Health, 10000 Zagreb, Croatia
| | | | - Thomas Ferenc
- Clinical Department of Diagnostic and Interventional Radiology, Merkur University Hospital, 10000 Zagreb, Croatia
| | - Irena Tabain
- Department of Virology, Croatian Institute of Public Health, 10000 Zagreb, Croatia
| | - Ljubo Barbic
- Department of Microbiology and Infectious Diseases with Clinic, Faculty of Veterinary Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Mateja Vujica Ferenc
- Department of Obstetrics and Gynecology, University Hospital Center Zagreb, 10000 Zagreb, Croatia
| | - Ivana Ferencak
- Department of Virology, Croatian Institute of Public Health, 10000 Zagreb, Croatia
| | - Vladimir Stevanovic
- Department of Microbiology and Infectious Diseases with Clinic, Faculty of Veterinary Medicine, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
13
|
Frasca L, Ocone G, Palazzo R. Safety of COVID-19 Vaccines in Patients with Autoimmune Diseases, in Patients with Cardiac Issues, and in the Healthy Population. Pathogens 2023; 12:pathogens12020233. [PMID: 36839505 PMCID: PMC9964607 DOI: 10.3390/pathogens12020233] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/25/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) has been a challenge for the whole world since the beginning of 2020, and COVID-19 vaccines were considered crucial for disease eradication. Instead of producing classic vaccines, some companies pointed to develop products that mainly function by inducing, into the host, the production of the antigenic protein of SARS-CoV-2 called Spike, injecting an instruction based on RNA or a DNA sequence. Here, we aim to give an overview of the safety profile and the actual known adverse effects of these products in relationship with their mechanism of action. We discuss the use and safety of these products in at-risk people, especially those with autoimmune diseases or with previously reported myocarditis, but also in the general population. We debate the real necessity of administering these products with unclear long-term effects to at-risk people with autoimmune conditions, as well as to healthy people, at the time of omicron variants. This, considering the existence of therapeutic interventions, much more clearly assessed at present compared to the past, and the relatively lower aggressive nature of the new viral variants.
Collapse
|
14
|
AT1 receptor autoantibodies mediate effects of metabolic syndrome on dopaminergic vulnerability. Brain Behav Immun 2023; 108:255-268. [PMID: 36535607 DOI: 10.1016/j.bbi.2022.12.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/20/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
The metabolic syndrome has been associated to chronic peripheral inflammation and related with neuroinflammation and neurodegeneration, including Parkinson's disease. However, the responsible mechanisms are unclear. Previous studies have involved the brain renin-angiotensin system in progression of Parkinson's disease and the angiotensin receptor type 1 (AT1) has been recently revealed as a major marker of dopaminergic vulnerability in humans. Dysregulation of tissue renin-angiotensin system is a key common mechanism for all major components of metabolic syndrome. Circulating AT1 agonistic autoantibodies have been observed in several inflammation-related peripheral processes, and activation of AT1 receptors of endothelial cells, dopaminergic neurons and glial cells have been observed to disrupt endothelial blood -brain barrier and induce neurodegeneration, respectively. Using a rat model, we observed that metabolic syndrome induces overactivity of nigral pro-inflammatory renin-angiotensin system axis, leading to increase in oxidative stress and neuroinflammation and enhancing dopaminergic neurodegeneration, which was inhibited by treatment with AT1 receptor blockers (ARBs). In rats, metabolic syndrome induced the increase in circulating levels of LIGHT and other major pro-inflammatory cytokines, and 27-hydroxycholesterol. Furthermore, the rats showed a significant increase in serum levels of proinflammatory AT1 and angiotensin converting enzyme 2 (ACE2) autoantibodies, which correlated with levels of several metabolic syndrome parameters. We also found AT1 and ACE2 autoantibodies in the CSF of these rats. Effects of circulating autoantibodies were confirmed by chronic infusion of AT1 autoantibodies, which induced blood-brain barrier disruption, an increase in the pro-inflammatory renin-angiotensin system activity in the substantia nigra and a significant enhancement in dopaminergic neuron death in two different rat models of Parkinson's disease. Observations in the rat models, were analyzed in a cohort of parkinsonian and non-parkinsonian patients with or without metabolic syndrome. Non-parkinsonian patients with metabolic syndrome showed significantly higher levels of AT1 autoantibodies than non-parkinsonian patients without metabolic syndrome. However, there was no significant difference between parkinsonian patients with metabolic syndrome or without metabolic syndrome, which showed higher levels of AT1 autoantibodies than non-parkinsonian controls. This is consistent with our recent studies, showing significant increase of AT1 and ACE2 autoantibodies in parkinsonian patients, which was related to dopaminergic degeneration and neuroinflammation. Altogether may lead to a vicious circle enhancing the progression of the disease that may be inhibited by strategies against production of these autoantibodies or AT1 receptor blockers (ARBs).
Collapse
|
15
|
Autoimmune complications of COVID-19 and potential consequences for long-lasting disease syndromes. Transfus Apher Sci 2023; 62:103625. [PMID: 36585276 PMCID: PMC9757887 DOI: 10.1016/j.transci.2022.103625] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The latest WHO report determined the increasing diversity within the CoV-2 omicron and its descendent lineages. Some heavily mutated offshoots of BA.5 and BA.2, such as BA.4.6, BF.7, BQ.1.1, and BA.2.75, are responsible for about 20% of infections and are spreading rapidly in multiple countries. It is a sign that Omicron subvariants are now developing a capacity to be more immune escaping and may contribute to a new wave of COVID-19. Covid-19 infections often induce many alterations in human physiological defense and the natural control systems, with exacerbated activation of the inflammatory and homeostatic response, as for any infectious diseases. Severe activation of the early phase of hemostatic components, often occurs, leading to thrombotic complications and often contributing to a lethal outcome selectively in certain populations. Development of autoimmune complications increases the disease burden and lowers its prognosis. While the true mechanism still remains unclear, it is believed to mainly be related to the host autoimmune responses as demonstrated, only in some patients suffering from the presence of autoantibodies that worsens the disease evolution. In fact in some studies the development of autoantibodies to angiotensin converting enzyme 2 (ACE2) was identified, and in other studies autoantibodies, thought to be targeting interferon or binding to annexin A1, or autoantibodies to phospholipids were seen. Moreover, the occurrence of autoimmune heparin induced thrombocytopenia has also been described in infected patients treated with heparin for controlling thrombogenicity. This commentary focuses on the presence of various autoantibodies reported so far in Covid-19 diseases, exploring their association with the disease course and the durability of some related symptoms. Attempts are also made to further analyze the potential mechanism of actions and link the presence of antibodies with pathological complications.
Collapse
|
16
|
Brock S, Jackson DB, Soldatos TG, Hornischer K, Schäfer A, Diella F, Emmert MY, Hoerstrup SP. Whole patient knowledge modeling of COVID-19 symptomatology reveals common molecular mechanisms. FRONTIERS IN MOLECULAR MEDICINE 2023; 2:1035290. [PMID: 39086962 PMCID: PMC11285600 DOI: 10.3389/fmmed.2022.1035290] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/12/2022] [Indexed: 08/02/2024]
Abstract
Infection with SARS-CoV-2 coronavirus causes systemic, multi-faceted COVID-19 disease. However, knowledge connecting its intricate clinical manifestations with molecular mechanisms remains fragmented. Deciphering the molecular basis of COVID-19 at the whole-patient level is paramount to the development of effective therapeutic approaches. With this goal in mind, we followed an iterative, expert-driven process to compile data published prior to and during the early stages of the pandemic into a comprehensive COVID-19 knowledge model. Recent updates to this model have also validated multiple earlier predictions, suggesting the importance of such knowledge frameworks in hypothesis generation and testing. Overall, our findings suggest that SARS-CoV-2 perturbs several specific mechanisms, unleashing a pathogenesis spectrum, ranging from "a perfect storm" triggered by acute hyper-inflammation, to accelerated aging in protracted "long COVID-19" syndromes. In this work, we shortly report on these findings that we share with the community via 1) a synopsis of key evidence associating COVID-19 symptoms and plausible mechanisms, with details presented within 2) the accompanying "COVID-19 Explorer" webserver, developed specifically for this purpose (found at https://covid19.molecularhealth.com). We anticipate that our model will continue to facilitate clinico-molecular insights across organ systems together with hypothesis generation for the testing of potential repurposing drug candidates, new pharmacological targets and clinically relevant biomarkers. Our work suggests that whole patient knowledge models of human disease can potentially expedite the development of new therapeutic strategies and support evidence-driven clinical hypothesis generation and decision making.
Collapse
Affiliation(s)
| | | | - Theodoros G. Soldatos
- Molecular Health GmbH, Heidelberg, Germany
- SRH Hochschule, University of Applied Science, Heidelberg, Germany
| | | | | | | | - Maximilian Y. Emmert
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Wyss Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
- Department of Cardiothoracic and Vascular Surgery, German Heart Institute Berlin, Berlin, Germany
- Department of Cardiovascular Surgery, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Simon P. Hoerstrup
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Wyss Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
17
|
Hallmann E, Sikora D, Poniedziałek B, Szymański K, Kondratiuk K, Żurawski J, Brydak L, Rzymski P. IgG autoantibodies against ACE2 in SARS-CoV-2 infected patients. J Med Virol 2023; 95:e28273. [PMID: 36324055 PMCID: PMC9877908 DOI: 10.1002/jmv.28273] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/26/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022]
Abstract
How frequently autoantibodies against angiotensin-converting enzyme 2 (ACE2) occur in patients infected by SARS-CoV-2 is understudied and limited to investigations on a small sample size. The presence of these antibodies may contribute to the long-lasting effects of COVID-19 observed in some individuals, particularly if IgG-class antibodies would emerge in patients. This study assessed the prevalence of IgG autoantibodies against ACE2 in 1139 patients infected with SARS-CoV-2 and examined their relationship with severity, demographic characteristics, and status of vaccination against influenza. The overall prevalence of anti-ACE IgG antibodies in our cohort was 1.5%. Most of these individuals were men (76.5%) and underwent mild COVID-19, but some severe and asymptomatic cases were also observed. Patients with severe infection had twofold higher titers than mild and asymptomatic cases. Age, comorbidities, and influenza vaccination status were not related to antibody prevalence. The prevalence of IgG anti-SARS-CoV-2 antibodies (against nucleocapsid protein and S2 subunit, but not against receptor-binding domain) was higher in the subset with ACE2 autoantibodies. Further research is required to understand the potential spectrum and duration of effects of IgG autoantibodies against ACE2 in patients after SARS-CoV-2 infection, particularly concerning long COVID-19.
Collapse
Affiliation(s)
- Ewelina Hallmann
- Department of Influenza ResearchNational Influenza Center at the National Institute of Public Health NIH ‐ National Research Institute in WarsawWarsawPoland
| | - Dominika Sikora
- Department of Environmental MedicinePoznań University of Medical SciencesPoznanPoland,Doctoral SchoolPoznan University of Medical SciencesPoznanPoland
| | - Barbara Poniedziałek
- Department of Environmental MedicinePoznań University of Medical SciencesPoznanPoland
| | - Karol Szymański
- Department of Influenza ResearchNational Influenza Center at the National Institute of Public Health NIH ‐ National Research Institute in WarsawWarsawPoland
| | - Katarzyna Kondratiuk
- Department of Influenza ResearchNational Influenza Center at the National Institute of Public Health NIH ‐ National Research Institute in WarsawWarsawPoland
| | - Jakub Żurawski
- Department of ImmunobiologyPoznan University of Medical SciencesPoznanPoland
| | - Lidia Brydak
- Department of Influenza ResearchNational Influenza Center at the National Institute of Public Health NIH ‐ National Research Institute in WarsawWarsawPoland
| | - Piotr Rzymski
- Department of Environmental MedicinePoznań University of Medical SciencesPoznanPoland,Integrated Science AssociationUniversal Scientific Education and Research NetworkPoznanPoland
| |
Collapse
|
18
|
Chao CH, Cheng D, Huang SW, Chuang YC, Yeh TM, Wang JR. Serological responses triggered by different SARS-CoV-2 vaccines against SARS-CoV-2 variants in Taiwan. Front Immunol 2022; 13:1023943. [PMID: 36458016 PMCID: PMC9705976 DOI: 10.3389/fimmu.2022.1023943] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 10/31/2022] [Indexed: 09/05/2023] Open
Abstract
Broadly neutralizing ability is critical for developing the next-generation SARS-CoV-2 vaccine. We collected sera samples between December 2021-January 2022 from 113 Taiwan naïve participants after their second dose of homologous vaccine (AZD1222, mRNA-1273, BNT162-b2, and MVC-COV1901) and compared the differences in serological responses of various SARS-CoV-2 vaccines. Compared to AZD1222, the two mRNA vaccines could elicit a higher level of anti-S1-RBD binding antibodies with higher broadly neutralizing ability evaluated using pseudoviruses of various SARS-CoV-2 lineages. The antigenic maps produced from the neutralization data implied that Omicron represents very different antigenic characteristics from the ancestral lineage. These results suggested that constantly administering the vaccine with ancestral Wuhan spike is insufficient for the Omicron outbreak. In addition, we found that anti-ACE2 autoantibodies were significantly increased in all four vaccinated groups compared to the unvaccinated pre-pandemic group, which needed to be investigated in the future.
Collapse
Affiliation(s)
- Chiao-Hsuan Chao
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Dayna Cheng
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Sheng-Wen Huang
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Tainan, Taiwan
| | | | - Trai-Ming Yeh
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
| | - Jen-Ren Wang
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan, Taiwan
| |
Collapse
|
19
|
Briquez PS, Rouhani SJ, Yu J, Pyzer AR, Trujillo J, Dugan HL, Stamper CT, Changrob S, Sperling AI, Wilson PC, Gajewski TF, Hubbell JA, Swartz MA. Severe COVID-19 induces autoantibodies against angiotensin II that correlate with blood pressure dysregulation and disease severity. SCIENCE ADVANCES 2022; 8:eabn3777. [PMID: 36206332 PMCID: PMC9544317 DOI: 10.1126/sciadv.abn3777] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 08/24/2022] [Indexed: 05/26/2023]
Abstract
Patients infected with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can experience life-threatening respiratory distress, blood pressure dysregulation, and thrombosis. This is thought to be associated with an impaired activity of angiotensin-converting enzyme 2 (ACE2), which is the main entry receptor of SARS-CoV-2 and which also tightly regulates blood pressure by converting the vasoconstrictive peptide angiotensin II (AngII) to a vasopressor peptide. Here, we show that a significant proportion of hospitalized patients with COVID-19 developed autoantibodies against AngII, whose presence correlates with lower blood oxygenation, blood pressure dysregulation, and overall higher disease severity. Anti-AngII antibodies can develop upon specific immune reaction to the SARS-CoV-2 proteins Spike or receptor-binding domain (RBD), to which they can cross-bind, suggesting some epitope mimicry between AngII and Spike/RBD. These results provide important insights on how an immune reaction against SARS-CoV-2 can impair blood pressure regulation.
Collapse
Affiliation(s)
- Priscilla S. Briquez
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
- Department of General and Visceral Surgery, Medical Center–University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany
| | - Sherin J. Rouhani
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Jovian Yu
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Athalia R. Pyzer
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Jonathan Trujillo
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Haley L. Dugan
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Christopher T. Stamper
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Siriruk Changrob
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL, USA
| | - Anne I. Sperling
- Committee on Immunology, University of Chicago, Chicago, IL, USA
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, IL, USA
- Department of Medicine, Department of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, VA, USA
| | - Patrick C. Wilson
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Thomas F. Gajewski
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
- Ben May Department of Cancer Research, University of Chicago, Chicago, IL, USA
- Committee on Cancer Biology, University of Chicago, Chicago, IL, USA
| | - Jeffrey A. Hubbell
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
- Committee on Cancer Biology, University of Chicago, Chicago, IL, USA
| | - Melody A. Swartz
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
- Ben May Department of Cancer Research, University of Chicago, Chicago, IL, USA
- Committee on Cancer Biology, University of Chicago, Chicago, IL, USA
| |
Collapse
|
20
|
Jeremiah SS, Miyakawa K, Ryo A. Detecting SARS-CoV-2 neutralizing immunity: highlighting the potential of split nanoluciferase technology. J Mol Cell Biol 2022; 14:mjac023. [PMID: 35416249 PMCID: PMC9387144 DOI: 10.1093/jmcb/mjac023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/22/2022] [Accepted: 03/01/2022] [Indexed: 11/24/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has progressed over 2 years since its onset causing significant health concerns all over the world and is currently curtailed by mass vaccination. Immunity acquired against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can be following either infection or vaccination. However, one can never be sure whether the acquired immunity is adequate to protect the individual from subsequent infection because of three important factors: individual variations in humoral response dynamics, waning of protective antibodies over time, and the emergence of immune escape mutants. Therefore, a test that can accurately differentiate the protected from the vulnerable is the need of the hour. The plaque reduction neutralization assay is the conventional gold standard test for estimating the titers of neutralizing antibodies that confer protection. However, it has got several drawbacks, which hinder the practical application of this test for wide-scale usage. Hence, various tests have been developed to detect protective immunity against SARS-CoV-2 that directly or indirectly assess the presence of neutralizing antibodies to SARS-CoV-2 in a lower biosafety setting. In this review, the pros and cons of the currently available assays are elaborated in detail and special focus is put on the scope of the novel split nanoluciferase technology for detecting SARS-CoV-2 neutralizing antibodies.
Collapse
Affiliation(s)
| | - Kei Miyakawa
- Department of Microbiology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | - Akihide Ryo
- Department of Microbiology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| |
Collapse
|
21
|
Khajeh Pour S, Scoville C, Tavernier SS, Aghazadeh-Habashi A. Plasma angiotensin peptides as biomarkers of rheumatoid arthritis are correlated with anti-ACE2 auto-antibodies level and disease intensity. Inflammopharmacology 2022; 30:1295-1302. [PMID: 35618976 PMCID: PMC9134980 DOI: 10.1007/s10787-022-01008-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/06/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND This study aimed to explore a correlation between plasma angiotensin II/(1-7) (Ang II/Ang-(1-7)) ratio, anti-ACE2 autoantibodies level and disease activity in rheumatoid arthritis (RA) patients. METHODS In a pilot study, the plasma level of Ang II, Ang-(1-7), and anti-ACE2 autoantibodies of twelve RA patients (five in active stage and seven in remission) were measured using an LC-MS/MS method and an ELISA kit, respectively. RESULTS The Ang-(1-7) level was significantly higher in the remission group than in the active RA patients (7.63 ± 2.61 vs. 1.29 ± 0.81 ng/mL). On the contrary, the Ang II level was higher in those with active RA compared to the remission group (5.43 ± 1.82 vs. 0.87 ± 0.16 ng/mL). The mean ELISA score of anti-ACE2 autoantibodies in patients with active RA was significantly higher than patients in remission (1.41 ± 0.11 vs. 1.81 ± 0.11, p < 0.05). CONCLUSION This study result suggests that the angiotensin peptides concentration and anti-ACE2 autoantibodies levels can be used as biomarkers of RA. This will help clinicians evaluate better treatment success rates and disease prognosis to prevent long-term complications of RA.
Collapse
Affiliation(s)
| | - Craig Scoville
- Institute of Arthritis Research LLC, Idaho Falls, ID, USA
| | | | - Ali Aghazadeh-Habashi
- College of Pharmacy, Idaho State University, Pocatello, ID, USA.
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Idaho State University, Leonard Hall 212, Pocatello, ID, 83209-8288, USA.
| |
Collapse
|
22
|
Labandeira CM, Pedrosa MA, Quijano A, Valenzuela R, Garrido-Gil P, Sanchez-Andrade M, Suarez-Quintanilla JA, Rodriguez-Perez AI, Labandeira-Garcia JL. Angiotensin type-1 receptor and ACE2 autoantibodies in Parkinson´s disease. NPJ Parkinsons Dis 2022; 8:76. [PMID: 35701430 PMCID: PMC9198025 DOI: 10.1038/s41531-022-00340-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/26/2022] [Indexed: 11/09/2022] Open
Abstract
The role of autoimmunity in neurodegeneration has been increasingly suggested. The renin-angiotensin system (RAS) autoantibodies play a major role in several peripheral inflammatory processes. Dysregulation of brain RAS has been involved in neuroinflammation and neurodegeneration. We aimed to know whether angiotensin type-1 receptor (AT1) autoantibodies (AT1 agonists) and angiotensin-converting enzyme 2 (ACE2) autoantibodies (ACE2 antagonists) may be involved in Parkinson's disease (PD) progression and constitute a new therapeutical target. Both AT1 and ACE2 serum autoantibodies were higher in a group of 117 PD patients than in a group of 106 controls. Serum AT1 autoantibodies correlated with several cytokines, particularly Tumor Necrosis Factor Ligand Superfamily Member 14 (TNFSF14, LIGHT), and 27-hydroxycholesterol levels. Serum ACE2 autoantibodies correlated with AT1 autoantibodies. Both autoantibodies were found in cerebrospinal fluid (CSF) of four PD patients with CSF samples. Consistent with the observations in patients, experimental dopaminergic degeneration, induced by 6-hydroxydopamine, increased levels of autoantibodies in serum and CSF in rats, as well as LIGHT levels and transglutaminase activity in rat substantia nigra. In cultures, administration of AT1 autoantibodies enhanced dopaminergic neuron degeneration and increased levels of neuroinflammation markers, which was inhibited by the AT1 antagonist candesartan. The results suggest dysregulation of RAS autoantibodies as a new mechanism that can contribute to PD progression. Therapeutical strategies blocking the production, or the effects of these autoantibodies may be useful for PD treatment, and the results further support repurposing AT1 blockers (ARBs) as treatment against PD progression.
Collapse
Affiliation(s)
- Carmen M Labandeira
- Research Center for Molecular Medicine and Chronic diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain.,Neurology Service, Hospital Alvaro Cunqueiro, University Hospital Complex, Vigo, Spain. Neurology Service. University Hospital of Ourense, Ourense, Spain
| | - Maria A Pedrosa
- Research Center for Molecular Medicine and Chronic diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Aloia Quijano
- Research Center for Molecular Medicine and Chronic diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Rita Valenzuela
- Research Center for Molecular Medicine and Chronic diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Pablo Garrido-Gil
- Research Center for Molecular Medicine and Chronic diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Mariña Sanchez-Andrade
- Obstetric Service, University Clinical Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | | | - Ana I Rodriguez-Perez
- Research Center for Molecular Medicine and Chronic diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain. .,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| | - Jose L Labandeira-Garcia
- Research Center for Molecular Medicine and Chronic diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain. .,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| |
Collapse
|
23
|
Casciola-Rosen L, Thiemann DR, Andrade F, Trejo-Zambrano MI, Leonard EK, Spangler JB, Skinner NE, Bailey J, Yegnasubramanian S, Wang R, Vaghasia AM, Gupta A, Cox AL, Ray SC, Linville RM, Guo Z, Searson PC, Machamer CE, Desiderio S, Sauer LM, Laeyendecker O, Garibaldi BT, Gao L, Damarla M, Hassoun PM, Hooper JE, Mecoli CA, Christopher-Stine L, Gutierrez-Alamillo L, Yang Q, Hines D, Clarke WA, Rothman RE, Pekosz A, Fenstermacher KZ, Wang Z, Zeger SL, Rosen A. IgM anti-ACE2 autoantibodies in severe COVID-19 activate complement and perturb vascular endothelial function. JCI Insight 2022; 7:e158362. [PMID: 35349483 PMCID: PMC9090251 DOI: 10.1172/jci.insight.158362] [Citation(s) in RCA: 16] [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: 01/11/2022] [Accepted: 03/24/2022] [Indexed: 12/15/2022] Open
Abstract
BackgroundSome clinical features of severe COVID-19 represent blood vessel damage induced by activation of host immune responses initiated by the coronavirus SARS-CoV-2. We hypothesized autoantibodies against angiotensin-converting enzyme 2 (ACE2), the SARS-CoV-2 receptor expressed on vascular endothelium, are generated during COVID-19 and are of mechanistic importance.MethodsIn an opportunity sample of 118 COVID-19 inpatients, autoantibodies recognizing ACE2 were detected by ELISA. Binding properties of anti-ACE2 IgM were analyzed via biolayer interferometry. Effects of anti-ACE2 IgM on complement activation and endothelial function were demonstrated in a tissue-engineered pulmonary microvessel model.ResultsAnti-ACE2 IgM (not IgG) autoantibodies were associated with severe COVID-19 and found in 18/66 (27.2%) patients with severe disease compared with 2/52 (3.8%) of patients with moderate disease (OR 9.38, 95% CI 2.38-42.0; P = 0.0009). Anti-ACE2 IgM autoantibodies were rare (2/50) in non-COVID-19 ventilated patients with acute respiratory distress syndrome. Unexpectedly, ACE2-reactive IgM autoantibodies in COVID-19 did not undergo class-switching to IgG and had apparent KD values of 5.6-21.7 nM, indicating they are T cell independent. Anti-ACE2 IgMs activated complement and initiated complement-binding and functional changes in endothelial cells in microvessels, suggesting they contribute to the angiocentric pathology of COVID-19.ConclusionWe identify anti-ACE2 IgM as a mechanism-based biomarker strongly associated with severe clinical outcomes in SARS-CoV-2 infection, which has therapeutic implications.FUNDINGBill & Melinda Gates Foundation, Gates Philanthropy Partners, Donald B. and Dorothy L. Stabler Foundation, and Jerome L. Greene Foundation; NIH R01 AR073208, R01 AR069569, Institutional Research and Academic Career Development Award (5K12GM123914-03), National Heart, Lung, and Blood Institute R21HL145216, and Division of Intramural Research, National Institute of Allergy and Infectious Diseases; National Science Foundation Graduate Research Fellowship (DGE1746891).
Collapse
Affiliation(s)
| | | | | | | | - Elissa K. Leonard
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jamie B. Spangler
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, USA
- Translational Tissue Engineering Center
| | | | - Justin Bailey
- Department of Medicine, Division of Infectious Diseases; and
| | | | - Rulin Wang
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ajay M. Vaghasia
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Anuj Gupta
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrea L. Cox
- Department of Medicine, Division of Infectious Diseases; and
| | - Stuart C. Ray
- Department of Medicine, Division of Infectious Diseases; and
| | - Raleigh M. Linville
- Institute for NanoBioTechnology and
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Peter C. Searson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Institute for NanoBioTechnology and
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Stephen Desiderio
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lauren M. Sauer
- Adult Emergency Department, Johns Hopkins Hospital, Baltimore, Maryland, USA
- Johns Hopkins Biocontainment Unit, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Oliver Laeyendecker
- Department of Medicine, Division of Infectious Diseases; and
- Division of Intramural Medicine, National Institute of Allergy and Infectious Diseases, NIH, Baltimore, Maryland, USA
| | - Brian T. Garibaldi
- Johns Hopkins Biocontainment Unit, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
| | - Li Gao
- Department of Medicine, Division of Allergy and Clinical Immunology; and
| | - Mahendra Damarla
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
| | - Paul M. Hassoun
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
| | - Jody E. Hooper
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | - David Hines
- Department of Medicine, Division of Rheumatology
| | - William A. Clarke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Richard E. Rothman
- Adult Emergency Department, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Andrew Pekosz
- Department of Environmental Health and Engineering
- Department of Molecular Microbiology and Immunology, and
| | | | - Zitong Wang
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Scott L. Zeger
- Department of Medicine, Division of Rheumatology
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Antony Rosen
- Department of Medicine, Division of Rheumatology
- Department of Cell Biology and
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
24
|
Lai YC, Cheng YW, Chao CH, Chang YY, Chen CD, Tsai WJ, Wang S, Lin YS, Chang CP, Chuang WJ, Chen LY, Wang YR, Chang SY, Huang W, Wang JR, Tseng CK, Lin CK, Chuang YC, Yeh TM. Antigenic Cross-Reactivity Between SARS-CoV-2 S1-RBD and Its Receptor ACE2. Front Immunol 2022; 13:868724. [PMID: 35603169 PMCID: PMC9114768 DOI: 10.3389/fimmu.2022.868724] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/07/2022] [Indexed: 11/27/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an emerging virus responsible for the ongoing COVID-19 pandemic. SARS-CoV-2 binds to the human cell receptor angiotensin-converting enzyme 2 (ACE2) through its receptor-binding domain in the S1 subunit of the spike protein (S1-RBD). The serum levels of autoantibodies against ACE2 are significantly higher in patients with COVID-19 than in controls and are associated with disease severity. However, the mechanisms through which these anti-ACE2 antibodies are induced during SARS-CoV-2 infection are unclear. In this study, we confirmed the increase in antibodies against ACE2 in patients with COVID-19 and found a positive correlation between the amounts of antibodies against ACE2 and S1-RBD. Moreover, antibody binding to ACE2 was significantly decreased in the sera of some COVID-19 patients after preadsorption of the sera with S1-RBD, which indicated that antibodies against S1-RBD can cross-react with ACE2. To confirm this possibility, two monoclonal antibodies (mAbs 127 and 150) which could bind to both S1-RBD and ACE2 were isolated from S1-RBD-immunized mice. Measurement of the binding affinities by Biacore showed these two mAbs bind to ACE2 much weaker than binding to S1-RBD. Epitope mapping using synthetic overlapping peptides and hydrogen deuterium exchange mass spectrometry (HDX-MS) revealed that the amino acid residues P463, F464, E465, R466, D467 and E471 of S1-RBD are critical for the recognition by mAbs 127 and 150. In addition, Western blotting analysis showed that these mAbs could recognize ACE2 only in native but not denatured form, indicating the ACE2 epitopes recognized by these mAbs were conformation-dependent. The protein-protein interaction between ACE2 and the higher affinity mAb 127 was analyzed by HDX-MS and visualized by negative-stain transmission electron microscopy imaging combined with antigen-antibody docking. Together, our results suggest that ACE2-cross-reactive anti-S1-RBD antibodies can be induced during SARS-CoV-2 infection due to potential antigenic cross-reactivity between S1-RBD and its receptor ACE2.
Collapse
Affiliation(s)
- Yen-Chung Lai
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Leadgene Biomedical, Inc., Tainan, Taiwan
| | - Yu-Wei Cheng
- Leadgene Biomedical, Inc., Tainan, Taiwan
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chiao-Hsuan Chao
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | | | - Wei-Jiun Tsai
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shuying Wang
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yee-Shin Lin
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chih-Peng Chang
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Woei-Jer Chuang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | | | - Sui-Yuan Chang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wenya Huang
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jen-Ren Wang
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | | | - Yung-Chun Chuang
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Leadgene Biomedical, Inc., Tainan, Taiwan
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Trai-Ming Yeh
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
25
|
Matsunaga A, Tsuzuki S, Morioka S, Ohmagari N, Ishizaka Y. Long COVID: current status in Japan and knowledge about its molecular background. Glob Health Med 2022; 4:83-93. [PMID: 35586759 PMCID: PMC9066464 DOI: 10.35772/ghm.2022.01013] [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: 03/09/2022] [Revised: 03/23/2022] [Accepted: 04/04/2022] [Indexed: 06/15/2023]
Abstract
Even after recovering from coronavirus disease 2019 (COVID-19), patients can experience prolonged complaints, referred to as "long COVID". Similar to reports in Caucasians, a follow-up study in Japan revealed that fatigue, dyspnea, cough, anosmia/dysgeusia, and dyssomnia are common symptoms. Although the precise mode of long COVID remains elusive, multiple etiologies such as direct organ damage by infection with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), autoimmunity, prolonged inflammatory reactions, and psychiatric impairment seem to be involved. Notably, SARS-CoV-2 is neurotropic, and viral RNA and proteins are continuously detectable in multiple organs, including the brain. Viral proteins exert a number of different toxic effects on cells, suggesting that persistent infection is a key element for understanding long COVID. Here, we first reviewed the current status of long COVID in Japan, and then summarized literature that help us understand the molecular background of the symptoms. Finally, we discuss the feasibility of vaccination as a treatment for patients with long COVID.
Collapse
Affiliation(s)
- Akihiro Matsunaga
- Department of Intractable Diseases, National Center for Global Health and Medicine, Tokyo, Japan
| | - Shinya Tsuzuki
- AMR Clinical Reference Center, National Center for Global Health and Medicine Hospital, Tokyo, Japan
| | - Shinichiro Morioka
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Norio Ohmagari
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yukihito Ishizaka
- Department of Intractable Diseases, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
26
|
Labandeira-Garcia JL, Labandeira CM, Valenzuela R, Pedrosa MA, Quijano A, Rodriguez-Perez AI. Drugs Modulating Renin-Angiotensin System in COVID-19 Treatment. Biomedicines 2022; 10:502. [PMID: 35203711 PMCID: PMC8962306 DOI: 10.3390/biomedicines10020502] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 02/07/2023] Open
Abstract
A massive worldwide vaccination campaign constitutes the main tool against the COVID-19 pandemic. However, drug treatments are also necessary. Antivirals are the most frequently considered treatments. However, strategies targeting mechanisms involved in disease aggravation may also be effective. A major role of the tissue renin-angiotensin system (RAS) in the pathophysiology and severity of COVID-19 has been suggested. The main link between RAS and COVID-19 is angiotensin-converting enzyme 2 (ACE2), a central RAS component and the primary binding site for SARS-CoV-2 that facilitates the virus entry into host cells. An initial suggestion that the susceptibility to infection and disease severity may be enhanced by angiotensin type-1 receptor blockers (ARBs) and ACE inhibitors (ACEIs) because they increase ACE2 levels, led to the consideration of discontinuing treatments in thousands of patients. More recent experimental and clinical data indicate that ACEIs and, particularly, ARBs can be beneficial for COVID-19 outcome, both by reducing inflammatory responses and by triggering mechanisms (such as ADAM17 inhibition) counteracting viral entry. Strategies directly activating RAS anti-inflammatory components such as soluble ACE2, Angiotensin 1-7 analogues, and Mas or AT2 receptor agonists may also be beneficial. However, while ACEIs and ARBs are cheap and widely used, the second type of strategies are currently under study.
Collapse
Affiliation(s)
- Jose L. Labandeira-Garcia
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (C.M.L.); (R.V.); (M.A.P.); (A.Q.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
| | - Carmen M. Labandeira
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (C.M.L.); (R.V.); (M.A.P.); (A.Q.)
- Neurology Service, Hospital Alvaro Cunqueiro, University Hospital Complex, 36213 Vigo, Spain
| | - Rita Valenzuela
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (C.M.L.); (R.V.); (M.A.P.); (A.Q.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
| | - Maria A. Pedrosa
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (C.M.L.); (R.V.); (M.A.P.); (A.Q.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
| | - Aloia Quijano
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (C.M.L.); (R.V.); (M.A.P.); (A.Q.)
| | - Ana I. Rodriguez-Perez
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (C.M.L.); (R.V.); (M.A.P.); (A.Q.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
| |
Collapse
|
27
|
Soós B, Fagyas M, Horváth Á, Végh E, Pusztai A, Czókolyová M, Csongrádi A, Hamar A, Pethő Z, Bodnár N, Kerekes G, Hodosi K, Szekanecz É, Szamosi S, Szántó S, Szűcs G, Papp Z, Szekanecz Z. Angiotensin Converting Enzyme Activity in Anti-TNF-Treated Rheumatoid Arthritis and Ankylosing Spondylitis Patients. Front Med (Lausanne) 2022; 8:785744. [PMID: 35155468 PMCID: PMC8828652 DOI: 10.3389/fmed.2021.785744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/10/2021] [Indexed: 11/28/2022] Open
Abstract
Introduction Angiotensin-converting enzyme (ACE) and ACE2 have been implicated in the regulation of vascular physiology. Elevated synovial and decreased or normal ACE or ACE2 levels have been found in rheumatoid arthritis (RA). Very little is known about the effects of tumor necrosis factor α (TNF-α) inhibition on ACE or ACE2 homeostasis. In this study, we assessed the effects of one-year anti-TNF therapy on ACE and ACE2 production in RA and ankylosing spondylitis (AS) in association with other biomarkers. Patients and Methods Forty patients including 24 RA patients treated with either etanercept (ETN) or certolizumab pegol (CZP) and 16 AS patients treated with ETN were included in a 12-month follow-up study. Serum ACE levels were determined by commercial ELISA, while serum ACE2 activity was assessed using a specific quenched fluorescent substrate. Ultrasonography was performed to determine flow-mediated vasodilation (FMD), common carotid intima-media thickness (ccIMT) and arterial pulse-wave velocity (PWV) in all patients. In addition, CRP, rheumatoid factor (RF) and ACPA were also measured. All assessments were performed at baseline and 6 and 12 months after treatment initiation. Results Anti-TNF therapy increased ACE levels in the full cohort, as well as in the RA and AS subsets. ACE2 activity increased in the full cohort, while the ACE/ACE2 ratio increased in the full cohort and in the RA subset (p < 0.05). Uni- and multivariable regression analyses determined associations between ACE or ACE/ACE2 ratios at different time points and disease duration, CRP, RF, FMD and IMT (p < 0.05). ACE2 activity correlated with CRP. The changes of ACE or ACE2 over 12 months were determined by treatment together with either RF or FMD (p < 0.05). Conclusions Anti-TNF treatment may increase ACE and ACE2 in the sera of RA and AS patients. ACE and ACE2 may be associated with disease duration, markers of inflammation and vascular pathophysiology. The effects of TNF inhibition on ACE and ACE2 may reflect, in part, the effects of these biologics on the cardiovascular system.
Collapse
Affiliation(s)
- Boglárka Soós
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Miklós Fagyas
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ágnes Horváth
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Edit Végh
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Anita Pusztai
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Monika Czókolyová
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Alexandra Csongrádi
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Hamar
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsófia Pethő
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Nóra Bodnár
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - György Kerekes
- Intensive Care Unit, Department of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Katalin Hodosi
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Éva Szekanecz
- Department of Oncology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Szilvia Szamosi
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Sándor Szántó
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Department of Sports Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gabriella Szűcs
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Szekanecz
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
28
|
Simon MA, Hanrott K, Budd DC, Torres F, Grünig E, Escribano‐Subias P, Meseguer ML, Halank M, Opitz C, Hall DA, Hewens D, Powley WM, Siederer S, Bayliffe A, Lazaar AL, Cahn A, Rosenkranz S. An open‐label, dose‐escalation study to evaluate the safety, tolerability, pharmacokinetics, and pharmacodynamics of single doses of GSK2586881 in participants with pulmonary arterial hypertension. Pulm Circ 2022; 12:e12024. [PMID: 35506108 PMCID: PMC9053011 DOI: 10.1002/pul2.12024] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 01/13/2023] Open
Abstract
Preclinical and early clinical studies suggest that angiotensin‐converting enzyme type 2 activity may be impaired in patients with pulmonary arterial hypertension (PAH); therefore, administration of exogenous angiotensin‐converting enzyme type 2 (ACE2) may be beneficial. This Phase IIa, multi‐center, open‐label, exploratory, single‐dose, dose‐escalation study (NCT03177603) assessed the potential vasodilatory effects of single doses of GSK2586881 (a recombinant human ACE2) on acute cardiopulmonary hemodynamics in hemodynamically stable adults with documented PAH who were receiving background PAH therapy. Successive cohorts of participants were administered a single intravenous dose of GSK2586881 of 0.1, 0.2, 0.4, or 0.8 mg/kg. Dose escalation occurred after four or more participants per cohort were dosed and a review of safety, tolerability, pharmacokinetics, and hemodynamic data up to 24 h postdose was undertaken. The primary endpoint was a change in cardiopulmonary hemodynamics (pulmonary vascular resistance, cardiac index, and mean pulmonary artery pressure) from baseline. Secondary/exploratory objectives included safety and tolerability, effect on renin‐angiotensin system peptides, and pharmacokinetics. GSK2586881 demonstrated no consistent or sustained effect on acute cardiopulmonary hemodynamics in participants with PAH receiving background PAH therapy (N = 23). All doses of GSK2586881 were well tolerated. GSK2586881 was quantifiable in plasma for up to 4 h poststart of infusion in all participants and caused a consistent and sustained reduction in angiotensin II and a corresponding increase in angiotensin (1–7) and angiotensin (1–5). While there does not appear to be a consistent acute vasodilatory response to single doses of GSK2586881 in participants with PAH, the potential benefits in terms of chronic vascular remodeling remain to be determined.
Collapse
Affiliation(s)
- Marc A. Simon
- Division of Cardiology, Department of Medicine University of California San Francisco California USA
| | - Kate Hanrott
- Research and Development, Medicines Research Centre GlaxoSmithKline plc. Stevenage UK
| | - David C. Budd
- Research and Development, Medicines Research Centre GlaxoSmithKline plc. Stevenage UK
| | | | - Ekkehard Grünig
- Centre for Pulmonary Hypertension Thoraxklinik Heidelberg gGmbH at Heidelberg University Hospital Heidelberg Germany
| | - Pilar Escribano‐Subias
- CIBER‐CV Cardiology Department, Pulmonary Hypertension Unit Hospital Universitario 12 de Octubre Madrid Spain
| | - Manuel L. Meseguer
- Lung Transplant and Pulmonary Vascular Diseases Department Hospital Universitari Vall d'Hebron Barcelona Spain
| | - Michael Halank
- Department of Internal Medicine I University Hospital Carl Gustav Carus Dresden Germany
| | - Christian Opitz
- Department of Cardiology DRK Kliniken Berlin Germany
- Department of Cardiology, University Heart Center Berlin Charité University Medicine Berlin Germany
| | - David A. Hall
- Research and Development, Medicines Research Centre GlaxoSmithKline plc. Stevenage UK
| | - Deborah Hewens
- Research and Development, Medicines Research Centre GlaxoSmithKline plc. Stevenage UK
| | - William M. Powley
- Research and Development, Medicines Research Centre GlaxoSmithKline plc. Stevenage UK
| | - Sarah Siederer
- Research and Development, Medicines Research Centre GlaxoSmithKline plc. Stevenage UK
| | - Andrew Bayliffe
- Research and Development, Medicines Research Centre GlaxoSmithKline plc. Stevenage UK
- Marengo Therapeutics and Apple Tree Partners Cambridge Massachusetts USA
| | - Aili L. Lazaar
- Discovery Medicine, Clinical Pharmacology and Experimental Medicine GlaxoSmithKline plc. Collegeville Pennsylvania USA
| | - Anthony Cahn
- Research and Development, Medicines Research Centre GlaxoSmithKline plc. Stevenage UK
| | - Stephan Rosenkranz
- Department III of Internal Medicine, Cologne Cardiovascular Research Center (CCRC) Cologne University Heart Center Cologne Germany
| |
Collapse
|
29
|
Amezcua-Guerra LM, Del Valle L, González-Pacheco H, Springall R, Márquez-Velasco R, Massó F, Brianza-Padilla M, Manzur-Sandoval D, González-Flores J, García-Ávila C, Juárez-Vicuña Y, Sánchez-Muñoz F, Ballinas-Verdugo MA, Basilio-Gálvez E, Paez-Arenas A, Castillo-Salazar M, Cásares-Alvarado S, Hernández-Diazcouder A, Sánchez-Gloria JL, Tavera-Alonso C, Gopar-Nieto R, Sandoval J. The prognostic importance of the angiotensin II/angiotensin-(1-7) ratio in patients with SARS-CoV-2 infection. Ther Adv Respir Dis 2022; 16:17534666221122544. [PMID: 36082632 PMCID: PMC9465579 DOI: 10.1177/17534666221122544] [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] [Indexed: 11/29/2022] Open
Abstract
Background: Information about angiotensin II (Ang II), angiotensin-converting enzyme 2
(ACE2), and Ang-(1–7) levels in patients with COVID-19 is scarce. Objective: To characterize the Ang II–ACE2–Ang-(1–7) axis in patients with SARS-CoV-2
infection to understand its role in pathogenesis and prognosis. Methods: Patients greater than 18 years diagnosed with COVID-19, based on clinical
findings and positive RT-PCR test, who required hospitalization and
treatment were included. We compared Ang II, aldosterone, Ang-(1–7), and
Ang-(1–9) concentrations and ACE2 concentration and activity between
COVID-19 patients and historic controls. We compared baseline demographics,
laboratory results (enzyme, peptide, and inflammatory marker levels), and
outcome (patients who survived versus those who died). Results: Serum from 74 patients [age: 58 (48–67.2) years; 68% men] with moderate (20%)
or severe (80%) COVID-19 were analyzed. During 13 (10–21) days of
hospitalization, 25 patients died from COVID-19 and 49 patients survived.
Compared with controls, Ang II concentration was higher and Ang-(1–7)
concentration was lower, despite significantly higher ACE2 activity in
patients. Ang II concentration was higher and Ang-(1–7) concentration was
lower in patients who died. The Ang II/Ang-(1–7) ratio was significantly
higher in patients who died. In multivariate analysis, Ang II/Ang-(1–7)
ratio greater than 3.45 (OR = 5.87) and lymphocyte count
⩽0.65 × 103/µl (OR = 8.43) were independent predictors of
mortality from COVID-19. Conclusion: In patients with severe SARS-CoV-2 infection, imbalance in the Ang
II–ACE2–Ang-(1–7) axis may reflect deleterious effects of Ang II and may
indicate a worse outcome.
Collapse
Affiliation(s)
- Luis M Amezcua-Guerra
- Immunology Department, Ignacio Chávez National Institute of Cardiology, Mexico City, Mexico
| | - Leonardo Del Valle
- Pharmacology Department, Ignacio Chávez National Institute of Cardiology, Mexico City, Mexico
| | | | - Rashidi Springall
- Immunology Department, Ignacio Chávez National Institute of Cardiology, Mexico City, Mexico
| | | | - Felipe Massó
- Translational Medicine Lab UNAM-INC Unit, Ignacio Chávez National Institute of Cardiology, Mexico City, Mexico
| | | | - Daniel Manzur-Sandoval
- Intensive Care Unit, Ignacio Chávez National Institute of Cardiology, Mexico City, Mexico
| | | | - Carlos García-Ávila
- Immunology Department, Ignacio Chávez National Institute of Cardiology, Mexico City, Mexico
| | - Yaneli Juárez-Vicuña
- Immunology Department, Ignacio Chávez National Institute of Cardiology, Mexico City, Mexico
| | - Fausto Sánchez-Muñoz
- Immunology Department, Ignacio Chávez National Institute of Cardiology, Mexico City, Mexico
| | | | - Edna Basilio-Gálvez
- Immunology Department, Ignacio Chávez National Institute of Cardiology, Mexico City, Mexico
| | - Araceli Paez-Arenas
- Translational Medicine Lab UNAM-INC Unit, Ignacio Chávez National Institute of Cardiology, Mexico City, Mexico
| | | | | | | | - José L Sánchez-Gloria
- Immunology Department, Ignacio Chávez National Institute of Cardiology, Mexico City, Mexico
| | | | - Rodrigo Gopar-Nieto
- Coronary Care Unit, Ignacio Chávez National Institute of Cardiology, Mexico City, Mexico
| | - Julio Sandoval
- Immunology Department, Ignacio Chávez National Institute of Cardiology, Juan Badiano # 1, Colonia Sección XVI Tlalpan, México City 14080, México
| |
Collapse
|
30
|
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: 2.0] [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
|
31
|
Krut VG, Astrakhantseva IV, Chuvpilo SA, Efimov GA, Ambaryan SG, Drutskaya MS, Nedospasov SA. Antibodies to the N-Terminal Domain of Angiotensin-Converting Enzyme (ACE2) That Block Its Interaction with SARS-CoV-2 S Protein. DOKL BIOCHEM BIOPHYS 2021; 502:1-4. [PMID: 34874527 PMCID: PMC8649309 DOI: 10.1134/s160767292201001x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 10/15/2021] [Accepted: 10/15/2021] [Indexed: 11/23/2022]
Abstract
SARS-CoV-2 is a new coronavirus that is the cause of COVID-19 pandemic. To enter the cell, the virus interacts via its surface S protein with angiotensin-converting enzyme 2 (ACE2), the main entry receptor on the cell membrane. Most of protective antibodies, including those induced by vaccinations, target the S protein, preventing its interaction with the ACE2 receptor. We have evaluated an alternative strategy for blocking the S-ACE2 interaction using new antipeptide antibodies to the N-terminus of the ACE2 molecule. These antibodies allow detection of human ACE2 in vitro and ex vivo.
Collapse
Affiliation(s)
- V G Krut
- Sirius University of Science and Technology, Federal Territory Sirius/Sochi, Russia
| | - I V Astrakhantseva
- Sirius University of Science and Technology, Federal Territory Sirius/Sochi, Russia
| | - S A Chuvpilo
- Sirius University of Science and Technology, Federal Territory Sirius/Sochi, Russia
| | - G A Efimov
- National Medical Research Center of Hematology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - S G Ambaryan
- Sirius University of Science and Technology, Federal Territory Sirius/Sochi, Russia
| | - M S Drutskaya
- Sirius University of Science and Technology, Federal Territory Sirius/Sochi, Russia.,Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - S A Nedospasov
- Sirius University of Science and Technology, Federal Territory Sirius/Sochi, Russia. .,Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
32
|
Al-Gburi S, Beissert S, Günther C. Molecular mechanisms of vasculopathy and coagulopathy in COVID-19. Biol Chem 2021; 402:1505-1518. [PMID: 34657406 DOI: 10.1515/hsz-2021-0245] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 10/06/2021] [Indexed: 01/08/2023]
Abstract
COVID-19 primarily affects the respiratory system and may lead to severe systemic complications, such as acute respiratory distress syndrome (ARDS), multiple organ failure, cytokine storm, and thromboembolic events. Depending on the immune status of the affected individual early disease control can be reached by a robust type-I-interferon (type-I-IFN) response restricting viral replication. If type-I-IFN upregulation is impaired, patients develop severe COVID-19 that involves profound alveolitis, endothelitis, complement activation, recruitment of immune cells, as well as immunothrombosis. In patients with proper initial disease control there can be a second flare of type-I-IFN release leading to post-COVID manifestation such as chilblain-like lesions that are characterized by thrombosis of small vessels in addition to an inflammatory infiltrate resembling lupus erythematosus (LE). Mechanistically, SARS-CoV-2 invades pneumocytes and endothelial cells by acting on angiotensin-II-converting enzyme 2 (ACE2). It is hypothesized, that viral uptake might downregulate ACE2 bioavailability and enhance angiotensin-II-derived pro-inflammatory and pro-thrombotic state. Since ACE2 is encoded on the X chromosome these conditions might also be influenced by gender-specific regulation. Taken together, SARS-CoV-2 infection affects the vascular compartment leading to variable thrombogenic or inflammatory response depending on the individual immune response status.
Collapse
Affiliation(s)
- Suzan Al-Gburi
- University Hospital Carl Gustav Carus, Technical University of Dresden, Fetscherstr. 74, D-01307 Dresden, Germany
| | - Stefan Beissert
- University Hospital Carl Gustav Carus, Technical University of Dresden, Fetscherstr. 74, D-01307 Dresden, Germany
| | - Claudia Günther
- University Hospital Carl Gustav Carus, Technical University of Dresden, Fetscherstr. 74, D-01307 Dresden, Germany
| |
Collapse
|
33
|
Briquez PS, Rouhani SJ, Yu J, Pyzer AR, Trujillo J, Dugan HL, Stamper CT, Changrob S, Sperling AI, Wilson PC, Gajewski TF, Hubbell JA, Swartz MA. SARS-CoV-2 infection induces cross-reactive autoantibodies against angiotensin II. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.11.02.21265789. [PMID: 34751272 PMCID: PMC8575143 DOI: 10.1101/2021.11.02.21265789] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Patients infected with the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) can experience life-threatening respiratory distress, blood pressure dysregulation and thrombosis. This is thought to be associated with an impaired activity of angiotensin-converting enzyme-2 (ACE-2), which is the main entry receptor of SARS-CoV-2 and which also tightly regulates blood pressure by converting the vasoconstrictive peptide angiotensin II (AngII) to a vasopressor peptide. Here, we show that a significant proportion of hospitalized COVID-19 patients developed autoantibodies against AngII, whose presence correlates with lower blood oxygenation, blood pressure dysregulation, and overall higher disease severity. Anti-AngII antibodies can develop upon specific immune reaction to the SARS-CoV-2 proteins Spike or RBD, to which they can cross-bind, suggesting some epitope mimicry between AngII and Spike/RBD. These results provide important insights on how an immune reaction against SARS-CoV-2 can impair blood pressure regulation.
Collapse
Affiliation(s)
- Priscilla S Briquez
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, Illinois, USA
| | - Sherin J Rouhani
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, Illinois, USA
| | - Jovian Yu
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, Illinois, USA
| | - Athalia R Pyzer
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, Illinois, USA
| | - Jonathan Trujillo
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, Illinois, USA
| | - Haley L Dugan
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, Illinois, USA
- Committee on Immunology, University of Chicago, Chicago, Illinois, USA
| | - Christopher T Stamper
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, Illinois, USA
- Committee on Immunology, University of Chicago, Chicago, Illinois, USA
| | - Siriruk Changrob
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, Illinois, USA
| | - Anne I Sperling
- Committee on Immunology, University of Chicago, Chicago, Illinois, USA
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, Illinois, USA
| | - Patrick C Wilson
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, Illinois, USA
- Committee on Immunology, University of Chicago, Chicago, Illinois, USA
| | - Thomas F Gajewski
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, Illinois, USA
- Committee on Immunology, University of Chicago, Chicago, Illinois, USA
- Ben May Department of Cancer Research, University of Chicago, Chicago, Illinois, USA
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois, USA
| | - Jeffrey A Hubbell
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, Illinois, USA
- Committee on Immunology, University of Chicago, Chicago, Illinois, USA
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois, USA
| | - Melody A Swartz
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, Illinois, USA
- Committee on Immunology, University of Chicago, Chicago, Illinois, USA
- Ben May Department of Cancer Research, University of Chicago, Chicago, Illinois, USA
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
34
|
Chang SE, Feng A, Meng W, Apostolidis SA, Mack E, Artandi M, Barman L, Bennett K, Chakraborty S, Chang I, Cheung P, Chinthrajah S, Dhingra S, Do E, Finck A, Gaano A, Geßner R, Giannini HM, Gonzalez J, Greib S, Gündisch M, Hsu AR, Kuo A, Manohar M, Mao R, Neeli I, Neubauer A, Oniyide O, Powell AE, Puri R, Renz H, Schapiro J, Weidenbacher PA, Wittman R, Ahuja N, Chung HR, Jagannathan P, James JA, Kim PS, Meyer NJ, Nadeau KC, Radic M, Robinson WH, Singh U, Wang TT, Wherry EJ, Skevaki C, Luning Prak ET, Utz PJ. New-onset IgG autoantibodies in hospitalized patients with COVID-19. Nat Commun 2021; 12:5417. [PMID: 34521836 PMCID: PMC8440763 DOI: 10.1038/s41467-021-25509-3] [Citation(s) in RCA: 259] [Impact Index Per Article: 86.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 08/09/2021] [Indexed: 02/08/2023] Open
Abstract
COVID-19 is associated with a wide range of clinical manifestations, including autoimmune features and autoantibody production. Here we develop three protein arrays to measure IgG autoantibodies associated with connective tissue diseases, anti-cytokine antibodies, and anti-viral antibody responses in serum from 147 hospitalized COVID-19 patients. Autoantibodies are identified in approximately 50% of patients but in less than 15% of healthy controls. When present, autoantibodies largely target autoantigens associated with rare disorders such as myositis, systemic sclerosis and overlap syndromes. A subset of autoantibodies targeting traditional autoantigens or cytokines develop de novo following SARS-CoV-2 infection. Autoantibodies track with longitudinal development of IgG antibodies recognizing SARS-CoV-2 structural proteins and a subset of non-structural proteins, but not proteins from influenza, seasonal coronaviruses or other pathogenic viruses. We conclude that SARS-CoV-2 causes development of new-onset IgG autoantibodies in a significant proportion of hospitalized COVID-19 patients and are positively correlated with immune responses to SARS-CoV-2 proteins.
Collapse
Affiliation(s)
- Sarah Esther Chang
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Allan Feng
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Wenzhao Meng
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sokratis A Apostolidis
- Department of Medicine, Division of Rheumatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elisabeth Mack
- Department of Hematology, Oncology, Immunology, Philipps University Marburg, Marburg, Germany
| | - Maja Artandi
- Department of Medicine, Division of Primary Care and Population Health, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford CROWN Clinic, Stanford University School of Medicine, Stanford, CA, USA
| | - Linda Barman
- Department of Medicine, Division of Primary Care and Population Health, Stanford University School of Medicine, Stanford, CA, USA
| | - Kate Bennett
- Molecular Pathology and Imaging Core, Department of Medicine, Gastroenterology Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Saborni Chakraborty
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Iris Chang
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA, USA
| | - Peggie Cheung
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Sharon Chinthrajah
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA, USA
| | - Shaurya Dhingra
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Evan Do
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA, USA
| | - Amanda Finck
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew Gaano
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Reinhard Geßner
- Institute of Laboratory Medicine, Philipps University Marburg, Marburg, Germany
| | - Heather M Giannini
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Joyce Gonzalez
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarah Greib
- Institute of Laboratory Medicine, Philipps University Marburg, Marburg, Germany
| | - Margrit Gündisch
- Institute of Laboratory Medicine, Philipps University Marburg, Marburg, Germany
| | - Alex Ren Hsu
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Alex Kuo
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Monali Manohar
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Rong Mao
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Indira Neeli
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Andreas Neubauer
- Department of Hematology, Oncology, Immunology, Philipps University Marburg, Marburg, Germany
| | - Oluwatosin Oniyide
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Abigail E Powell
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- ChEM-H, Stanford University, Stanford, USA
| | - Rajan Puri
- Department of Medicine, Division of Primary Care and Population Health, Stanford University School of Medicine, Stanford, CA, USA
| | - Harald Renz
- Institute of Laboratory Medicine, Philipps University Marburg, Marburg, Germany
- Member of the Universities of Giessen and Marburg Lung Center (UGMLC), and the German Center for Lung Research (DZL), Giessen, Germany
| | - Jeffrey Schapiro
- TPMG Regional Reference Laboratory, Kaiser Permanente Northern California, Berkeley, CA, USA
| | - Payton A Weidenbacher
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- ChEM-H, Stanford University, Stanford, USA
| | - Richard Wittman
- Department of Medicine, Division of Primary Care and Population Health, Stanford University School of Medicine, Stanford, CA, USA
| | - Neera Ahuja
- Department of Medicine, Division of Hospital Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Ho-Ryun Chung
- Institute for Medical Bioinformatics and Biostatistics, Philipps University Marburg, Marburg, Germany
| | - Prasanna Jagannathan
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Judith A James
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Peter S Kim
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Nuala J Meyer
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kari C Nadeau
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA, USA
| | - Marko Radic
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - William H Robinson
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
- VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Upinder Singh
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Taia T Wang
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - E John Wherry
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chrysanthi Skevaki
- Institute of Laboratory Medicine, Philipps University Marburg, Marburg, Germany.
- Member of the Universities of Giessen and Marburg Lung Center (UGMLC), and the German Center for Lung Research (DZL), Giessen, Germany.
| | - Eline T Luning Prak
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Paul J Utz
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, USA.
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
35
|
Arthur JM, Forrest JC, Boehme KW, Kennedy JL, Owens S, Herzog C, Liu J, Harville TO. Development of ACE2 autoantibodies after SARS-CoV-2 infection. PLoS One 2021; 16:e0257016. [PMID: 34478478 PMCID: PMC8415618 DOI: 10.1371/journal.pone.0257016] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/20/2021] [Indexed: 12/31/2022] Open
Abstract
Background Activation of the immune system is implicated in the Post-Acute Sequelae after SARS-CoV-2 infection (PASC) but the mechanisms remain unknown. Angiotensin-converting enzyme 2 (ACE2) cleaves angiotensin II (Ang II) resulting in decreased activation of the AT1 receptor and decreased immune system activation. We hypothesized that autoantibodies against ACE2 may develop after SARS-CoV-2 infection, as anti-idiotypic antibodies to anti-spike protein antibodies. Methods and findings We tested plasma or serum for ACE2 antibodies in 67 patients with known SARS-CoV-2 infection and 13 with no history of infection. None of the 13 patients without history of SARS-CoV-2 infection and 1 of the 20 outpatients that had a positive PCR test for SARS-CoV-2 had levels of ACE2 antibodies above the cutoff threshold. In contrast, 26/32 (81%) in the convalescent group and 14/15 (93%) of patients acutely hospitalized had detectable ACE2 antibodies. Plasma from patients with antibodies against ACE2 had less soluble ACE2 activity in plasma but similar amounts of ACE2 protein compared to patients without ACE2 antibodies. We measured the capacity of the samples to inhibit ACE2 enzyme activity. Addition of plasma from patients with ACE2 antibodies led to decreased activity of an exogenous preparation of ACE2 compared to patients that did not have antibodies. Conclusions Many patients with a history of SARS-CoV-2 infection have antibodies specific for ACE2. Patients with ACE2 antibodies have lower activity of soluble ACE2 in plasma. Plasma from these patients also inhibits exogenous ACE2 activity. These findings are consistent with the hypothesis that ACE2 antibodies develop after SARS-CoV-2 infection and decrease ACE2 activity. This could lead to an increase in the abundance of Ang II, which causes a proinflammatory state that triggers symptoms of PASC.
Collapse
Affiliation(s)
- John M. Arthur
- Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
- Central Arkansas Veterans Healthcare System, Little Rock, AR, United States of America
- * E-mail:
| | - J. Craig Forrest
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
- Center for Microbial Pathogenesis and Host Inflammatory Responses, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Karl W. Boehme
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
- Center for Microbial Pathogenesis and Host Inflammatory Responses, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Joshua L. Kennedy
- Center for Microbial Pathogenesis and Host Inflammatory Responses, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Shana Owens
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Christian Herzog
- Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Juan Liu
- Department of Pathology and Laboratory Services, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Terry O. Harville
- Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
- Department of Pathology and Laboratory Services, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| |
Collapse
|
36
|
Amiral J, Busch MH, Timmermans SAMEG, Reutelingsperger CP, van Paassen P. Development of IgG, IgM, and IgA autoantibodies against angiotensin converting enzyme 2 in patients with COVID-19. J Appl Lab Med 2021; 7:382-386. [PMID: 34213568 DOI: 10.1093/jalm/jfab065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/13/2021] [Indexed: 11/13/2022]
Affiliation(s)
- Jean Amiral
- SH/Scientific-Hemostasis, Franconville, France
| | - Matthias H Busch
- Department of Nephrology and Clinical Immunology, Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands
| | - Sjoerd A M E G Timmermans
- Department of Nephrology and Clinical Immunology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Chris P Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands
| | - Pieter van Paassen
- Department of Nephrology and Clinical Immunology, Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands
| |
Collapse
|
37
|
Mohkhedkar M, Venigalla SSK, Janakiraman V. Untangling COVID-19 and autoimmunity: Identification of plausible targets suggests multi organ involvement. Mol Immunol 2021; 137:105-113. [PMID: 34242919 PMCID: PMC8241658 DOI: 10.1016/j.molimm.2021.06.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/03/2021] [Accepted: 06/27/2021] [Indexed: 10/28/2022]
Abstract
Underlying mechanisms of multi-organ manifestations and exacerbated inflammation in COVID-19 are yet to be delineated. The hypothesis of SARS-CoV-2 triggering autoimmunity is gaining attention and, in the present study, we have identified 28 human proteins harbouring regions homologous to SARS-CoV-2 peptides that could possibly be acting as autoantigens in COVID-19 patients displaying autoimmune conditions. Interestingly, these conserved regions are amongst the experimentally validated B cell epitopes of SARS-CoV-2 proteins. The reported human proteins have demonstrated presence of autoantibodies against them in typical autoimmune conditions which may explain the frequent occurrence of autoimmune conditions following SARS-CoV-2 infection. Moreover, the proposed autoantigens' widespread tissue distribution is suggestive of their involvement in multi-organ manifestations via molecular mimicry. We opine that our report may aid in directing subsequent necessary antigen-specific studies, results of which would be of long-term relevance in management of extrapulmonary symptoms of COVID-19.
Collapse
Affiliation(s)
- Mugdha Mohkhedkar
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - Siva Sai Krishna Venigalla
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - Vani Janakiraman
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India.
| |
Collapse
|
38
|
Rodriguez-Perez AI, Labandeira CM, Pedrosa MA, Valenzuela R, Suarez-Quintanilla JA, Cortes-Ayaso M, Mayán-Conesa P, Labandeira-Garcia JL. Autoantibodies against ACE2 and angiotensin type-1 receptors increase severity of COVID-19. J Autoimmun 2021; 122:102683. [PMID: 34144328 PMCID: PMC8193025 DOI: 10.1016/j.jaut.2021.102683] [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: 05/06/2021] [Revised: 05/30/2021] [Accepted: 06/01/2021] [Indexed: 02/07/2023]
Abstract
The renin-angiotensin system (RAS) plays a major role in COVID-19. Severity of several inflammation-related diseases has been associated with autoantibodies against RAS, particularly agonistic autoantibodies for angiotensin type-1 receptors (AA-AT1) and autoantibodies against ACE2 (AA-ACE2). Disease severity of COVID-19 patients was defined as mild, moderate or severe following the WHO Clinical Progression Scale and determined at medical discharge. Serum AA-AT1 and AA-ACE2 were measured in COVID-19 patients (n = 119) and non-infected controls (n = 23) using specific solid-phase, sandwich enzyme-linked immunosorbent assays. Serum LIGHT (TNFSF14; tumor necrosis factor ligand superfamily member 14) levels were measured with the corresponding assay kit. At diagnosis, AA-AT1 and AA-ACE2 levels were significantly higher in the COVID-19 group relative to controls, and we observed significant association between disease outcome and serum AA-AT1 and AA-ACE2 levels. Mild disease patients had significantly lower levels of AA-AT1 (p < 0.01) and AA-ACE2 (p < 0.001) than moderate and severe patients. No significant differences were detected between males and females. The increase in autoantibodies was not related to comorbidities potentially affecting COVID-19 severity. There was significant positive correlation between serum levels of AA-AT1 and LIGHT (TNFSF14; rPearson = 0.70, p < 0.001). Both AA-AT1 (by agonistic stimulation of AT1 receptors) and AA-ACE2 (by reducing conversion of Angiotensin II into Angiotensin 1-7) may lead to increase in AT1 receptor activity, enhance proinflammatory responses and severity of COVID-19 outcome. Patients with high levels of autoantibodies require more cautious control after diagnosis. Additionally, the results encourage further studies on the possible protective treatment with AT1 receptor blockers in COVID-19.
Collapse
Affiliation(s)
- Ana I Rodriguez-Perez
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Carmen M Labandeira
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain; Hospital Alvaro Cunqueiro, University Hospital Complex, Vigo, Spain
| | - Maria A Pedrosa
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Rita Valenzuela
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Juan A Suarez-Quintanilla
- Primary Health-Care Unit Fontiñas, IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - María Cortes-Ayaso
- Emergency Department, University Clinical Hospital of Santiago, Santiago de Compostela, Spain
| | - Placido Mayán-Conesa
- Emergency Department, University Clinical Hospital of Santiago, Santiago de Compostela, Spain
| | - Jose L Labandeira-Garcia
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain.
| |
Collapse
|
39
|
Bahouth MN, Venkatesan A. Acute Viral Illnesses and Ischemic Stroke: Pathophysiological Considerations in the Era of the COVID-19 Pandemic. Stroke 2021; 52:1885-1894. [PMID: 33794653 PMCID: PMC8078120 DOI: 10.1161/strokeaha.120.030630] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The severe acute respiratory syndrome coronavirus 2 or coronavirus disease 2019 (COVID-19) pandemic has raised concerns about the correlation with this viral illness and increased risk of stroke. Although it is too early in the pandemic to know the strength of the association between COVID-19 and stroke, it is an opportune time to review the relationship between acute viral illnesses and stroke. Here, we summarize pathophysiological principles and available literature to guide understanding of how viruses may contribute to ischemic stroke. After a review of inflammatory mechanisms, we summarize relevant pathophysiological principles of vasculopathy, hypercoagulability, and hemodynamic instability. We will end by discussing mechanisms by which several well-known viruses may cause stroke in an effort to inform our understanding of the relationship between COVID-19 and stroke.
Collapse
Affiliation(s)
- Mona N. Bahouth
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD
| | - Arun Venkatesan
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD
| |
Collapse
|
40
|
Miziołek B, Sieńczyk M, Grzywa R, Łupicka-Słowik A, Kucharz E, Kotyla P, Bergler-Czop B. The prevalence and role of functional autoantibodies to angiotensin-converting-enzyme-2 in patients with systemic sclerosis. Autoimmunity 2021; 54:181-186. [PMID: 33910447 DOI: 10.1080/08916934.2021.1916915] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Systemic sclerosis (SSc) is an autoimmune disease caused by the imbalance between the activity of angiotensin II and angiotensin-(1-7). Their balance should be controlled by angiotensin-converting enzyme 2 (ACE2), which degrades angiotensin II into angiotensin-(1-7). Previously, autoantibodies to ACE2 (anti-ACE2) were identified in patients with vasculopathy due to different connective tissue diseases, including SSc, but their frequency in SSc was not further analyzed. The aim of the research was to investigate the prevalence and potential role of those anti-ACE2 antibodies in SSc patients. MATERIALS AND METHODS There were enrolled 27 patients with SSc and 23 healthy donors. ELISA assay determined the presence of anti-ACE2 autoantibodies in serum samples. The results were compared to plasma measurements of angiotensin-(1-7) level via commercial ELISA. RESULTS The presence of anti-ACE2 autoantibodies was confirmed in five patients with SSc and two healthy controls. Two of those SSc subjects were anti-Scl70+, another two were double anti-Scl70+ and anti-Ro/SSA+, and anti-PM/Scl antibodies were detected in one patient. Median plasma level of Ang-(1-7) in anti-ACE2 negative patients was 47.4 pg/ml and stayed below the detection level in anti-ACE2 positive subjects. The plasma level of Ang-(1-7) was undetectable in four SSc patients, and three of them were anti-ACE2 positive. CONCLUSIONS Anti-ACE2 antibodies appear to be other functional autoantibodies with the potential to dysregulate the balance between Ang II and Ang-(1-7). They are non-specific for SSc and probably result from polyautoimmunity which affect some of SSc patients. Their occurrence in SSc settings may be associated with a severe depletion of plasma Ang-(1-7).
Collapse
Affiliation(s)
- Bartosz Miziołek
- Department of Dermatology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland.,Department of Internal Medicine Rheumatology and Clinical Immunology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Marcin Sieńczyk
- Department of Organic and Medicinal Chemistry, Faculty of Chemistry, Wroclaw University of Science and Technology, Wrocław, Poland
| | - Renata Grzywa
- Department of Organic and Medicinal Chemistry, Faculty of Chemistry, Wroclaw University of Science and Technology, Wrocław, Poland
| | - Agnieszka Łupicka-Słowik
- Department of Organic and Medicinal Chemistry, Faculty of Chemistry, Wroclaw University of Science and Technology, Wrocław, Poland
| | - Eugeniusz Kucharz
- Department of Internal Medicine Rheumatology and Clinical Immunology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Przemysław Kotyla
- Department of Internal Medicine Rheumatology and Clinical Immunology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Beata Bergler-Czop
- Department of Dermatology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
41
|
Zamai L. Upregulation of the Renin-Angiotensin System Pathways and SARS-CoV-2 Infection: The Rationale for the Administration of Zinc-Chelating Agents in COVID-19 Patients. Cells 2021; 10:506. [PMID: 33673459 PMCID: PMC7997276 DOI: 10.3390/cells10030506] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/12/2022] Open
Abstract
The article describes the rationale for the administration of zinc-chelating agents in COVID-19 patients. In a previous work I have highlighted that the binding of the SARS-CoV spike proteins to the zinc-metalloprotease ACE2 has been shown to induce ACE2 shedding by activating the zinc-metalloprotease ADAM17, which ultimately leads to systemic upregulation of ACE2 activity. Moreover, based on experimental models, it was also shown the detrimental effect of the excessive systemic activity of ACE2 through its downstream pathways, which leads to "clinical" manifestations resembling COVID-19. In this regard, strong upregulation of circulating ACE2 activity was recently reported in COVID-19 patients, thus supporting the previous hypothesis that COVID-19 may derive from upregulation of ACE2 activity. Based on this, a reasonable hypothesis of using inhibitors that curb the upregulation of both ACE2 and ADAM17 zinc-metalloprotease activities and consequent positive feedback-loops (initially triggered by SARS-CoV-2 and subsequently sustained independently on viral trigger) is proposed as therapy for COVID-19. In particular, zinc-chelating agents such as citrate and ethylenediaminetetraacetic acid (EDTA) alone or in combination are expected to act in protecting from COVID-19 at different levels thanks to their both anticoagulant properties and inhibitory activity on zinc-metalloproteases. Several arguments are presented in support of this hypothesis and based on the current knowledge of both beneficial/harmful effects and cost/effectiveness, the use of chelating agents in the prevention and therapy of COVID-19 is proposed. In this regard, clinical trials (currently absent) employing citrate/EDTA in COVID-19 are urgently needed in order to shed more light on the efficacy of zinc chelators against SARS-CoV-2 infection in vivo.
Collapse
Affiliation(s)
- Loris Zamai
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy;
- National Institute for Nuclear Physics (INFN)-Gran Sasso National Laboratory (LNGS), Assergi, 67100 L’Aquila, Italy
| |
Collapse
|
42
|
Chang SE, Feng A, Meng W, Apostolidis SA, Mack E, Artandi M, Barman L, Bennett K, Chakraborty S, Chang I, Cheung P, Chinthrajah S, Dhingra S, Do E, Finck A, Gaano A, Geßner R, Giannini HM, Gonzalez J, Greib S, Gündisch M, Hsu AR, Kuo A, Manohar M, Mao R, Neeli I, Neubauer A, Oniyide O, Powell AE, Puri R, Renz H, Schapiro JM, Weidenbacher PA, Wittman R, Ahuja N, Chung HR, Jagannathan P, James J, Kim PS, Meyer NJ, Nadeau K, Radic M, Robinson WH, Singh U, Wang TT, Wherry EJ, Skevaki C, Prak ETL, Utz PJ. New-Onset IgG Autoantibodies in Hospitalized Patients with COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021. [PMID: 33532787 DOI: 10.1101/2021.01.27.21250559] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Coronavirus Disease 2019 (COVID-19), caused by Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2), is associated with a wide range of clinical manifestations, including autoimmune features and autoantibody production. We developed three different protein arrays to measure hallmark IgG autoantibodies associated with Connective Tissue Diseases (CTDs), Anti-Cytokine Antibodies (ACA), and anti-viral antibody responses in 147 hospitalized COVID-19 patients in three different centers. Autoantibodies were identified in approximately 50% of patients, but in <15% of healthy controls. When present, autoantibodies largely targeted autoantigens associated with rare disorders such as myositis, systemic sclerosis and CTD overlap syndromes. Anti-nuclear antibodies (ANA) were observed in ∼25% of patients. Patients with autoantibodies tended to demonstrate one or a few specificities whereas ACA were even more prevalent, and patients often had antibodies to multiple cytokines. Rare patients were identified with IgG antibodies against angiotensin converting enzyme-2 (ACE-2). A subset of autoantibodies and ACA developed de novo following SARS-CoV-2 infection while others were transient. Autoantibodies tracked with longitudinal development of IgG antibodies that recognized SARS-CoV-2 structural proteins such as S1, S2, M, N and a subset of non-structural proteins, but not proteins from influenza, seasonal coronaviruses or other pathogenic viruses. COVID-19 patients with one or more autoantibodies tended to have higher levels of antibodies against SARS-CoV-2 Nonstructural Protein 1 (NSP1) and Methyltransferase (ME). We conclude that SARS-CoV-2 causes development of new-onset IgG autoantibodies in a significant proportion of hospitalized COVID-19 patients and are positively correlated with immune responses to SARS-CoV-2 proteins.
Collapse
|
43
|
Tang X, Geng L, Feng X, Sun L. Decreased serum ACE2 levels in patients with connective tissue diseases. Rheumatology (Oxford) 2020; 60:4401-4406. [PMID: 33369650 DOI: 10.1093/rheumatology/keaa898] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/24/2020] [Indexed: 11/14/2022] Open
Abstract
Abstract
Objective
To evaluate serum concentration and activity of angiotensin-converting enzyme 2 (ACE2) in patients with connective tissue diseases (CTDs).
Methods
Serum samples from healthy subjects and patients with SLE, systemic sclerosis (SSc), primary Sjögren’s syndrome (SS) and RA were collected. The concentration and activity of ACE2 were measured by ELISA and fluorometric method, respectively, and analysed for associations with clinical features and concurrent medications.
Results
In total, 66 SLE, 55 SSc, 31 SS and 31 RA patients were involved. ACE2 concentration was significantly decreased in patients with either of the four CTDs compared with healthy subjects. The concentration was not linked to special clinical features expect that it was slightly lower in patients with lupus nephritis than those without. In SLE patients, ACE2 concentration elevated with the increase of glucocorticoids, and was not associated with other treatments. Different from the concentration, ACE2 activity was increased in CTD patients. A weak correlation of ACE2 activity with SLE disease activity index score was also observed.
Conclusion
The clinical significance of ACE2 concentration and activity looks quite different among CTD patients. Preliminary data suggest ACE2 levels are not affected by most of the treatments.
Collapse
Affiliation(s)
- Xiaojun Tang
- Department of Rheumatology and Immunology, Nanjing University Medical School Affiliated Drum Tower Hospital, Nanjing, China
| | - Linyu Geng
- Department of Rheumatology and Immunology, Nanjing University Medical School Affiliated Drum Tower Hospital, Nanjing, China
| | - Xuebing Feng
- Department of Rheumatology and Immunology, Nanjing University Medical School Affiliated Drum Tower Hospital, Nanjing, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing University Medical School Affiliated Drum Tower Hospital, Nanjing, China
| |
Collapse
|
44
|
Meyer B, Reimerink J, Torriani G, Brouwer F, Godeke GJ, Yerly S, Hoogerwerf M, Vuilleumier N, Kaiser L, Eckerle I, Reusken C. Validation and clinical evaluation of a SARS-CoV-2 surrogate virus neutralisation test (sVNT). Emerg Microbes Infect 2020; 9:2394-2403. [PMID: 33043818 PMCID: PMC7605318 DOI: 10.1080/22221751.2020.1835448] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
To understand SARS-CoV-2 immunity after natural infection or vaccination, functional assays such as virus neutralising assays are needed. So far, assays to detect SARS-CoV-2 neutralising antibodies rely on cell-culture based infection assays either using wild type SARS-CoV-2 or pseudotyped viruses. Such assays are labour-intensive, require appropriate biosafety facilities and are difficult to standardize. Recently, a new surrogate virus neutralisation test (sVNT) was described that uses the principle of an ELISA to measure the neutralisation capacity of anti-SARS-CoV-2 antibodies directed against the receptor binding domain. Here, we performed an independent evaluation of the robustness, specificity and sensitivity on an extensive panel of sera from 269 PCR-confirmed COVID-19 cases and 259 unmatched samples collected before 2020 and compared it to cell-based neutralisation assays. We found a high specificity of 99.2 (95%CI: 96.9–99.9) and overall sensitivity of 80.3 (95%CI: 74.9–84.8) for the sVNT. Clinical sensitivity increased between early (<14 days post symptom onset or post diagnosis, dpos/dpd) and late sera (>14 dpos/dpd) from 75.0 (64.7–83.2) to 83.1 (76.5–88.1). Also, higher severity was associated with an increase in clinical sensitivity. Upon comparison with cell-based neutralisation assays we determined an analytical sensitivity of 74.3 (56.4–86.9) and 98.2 (89.4–99.9) for titres ≥10 to <40 and ≥40 to <160, respectively. Only samples with a titre ≥160 were always positive in the sVNT. In conclusion, the sVNT can be used as an additional assay to determine the immune status of COVID-19 infected of vaccinated individuals but its value needs to be assessed for each specific context.
Collapse
Affiliation(s)
- Benjamin Meyer
- Centre for Vaccinology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Johan Reimerink
- Centre for Infectious Disease Control, WHO COVID-19 reference laboratory, RIVM, Bilthoven, Netherlands
| | - Giulia Torriani
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Fion Brouwer
- Centre for Infectious Disease Control, WHO COVID-19 reference laboratory, RIVM, Bilthoven, Netherlands
| | - Gert-Jan Godeke
- Centre for Infectious Disease Control, WHO COVID-19 reference laboratory, RIVM, Bilthoven, Netherlands
| | - Sabine Yerly
- Laboratory of Virology, Geneva University Hospitals, Geneva, Switzerland
| | - Marieke Hoogerwerf
- Centre for Infectious Disease Control, WHO COVID-19 reference laboratory, RIVM, Bilthoven, Netherlands
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Department of Diagnostics, Geneva University Hospitals and Geneva University, Geneva, Switzerland.,Division of Laboratory Medicine, Department of Medicine, Faculty of Medicine, Geneva, Switzerland
| | - Laurent Kaiser
- Laboratory of Virology, Geneva University Hospitals, Geneva, Switzerland.,Division of Infectious Disease, Geneva University Hospitals, Geneva, Switzerland.,Geneva Centre for Emerging Viral Diseases, Geneva University Hospitals, Geneva, Switzerland
| | - Isabella Eckerle
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland.,Division of Infectious Disease, Geneva University Hospitals, Geneva, Switzerland.,Geneva Centre for Emerging Viral Diseases, Geneva University Hospitals, Geneva, Switzerland
| | - Chantal Reusken
- Centre for Infectious Disease Control, WHO COVID-19 reference laboratory, RIVM, Bilthoven, Netherlands
| |
Collapse
|
45
|
Giollo A, Cioffi G, Rossini M, Viapiana O, De Simone G. Very low reporting rate of connective tissue diseases among coronavirus disease 2019 (Covid-19) patients and the renin-angiotensin system - An overlooked association? Eur J Intern Med 2020; 80:106-107. [PMID: 32553588 PMCID: PMC7290229 DOI: 10.1016/j.ejim.2020.06.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/12/2020] [Accepted: 06/10/2020] [Indexed: 01/21/2023]
Affiliation(s)
- Alessandro Giollo
- Rheumatology Section, Department of Medicine, University of Verona, Verona, Italy.
| | - Giovanni Cioffi
- Rheumatology Section, Department of Medicine, University of Verona, Verona, Italy
| | - Maurizio Rossini
- Rheumatology Section, Department of Medicine, University of Verona, Verona, Italy
| | - Ombretta Viapiana
- Rheumatology Section, Department of Medicine, University of Verona, Verona, Italy
| | - Giovanni De Simone
- Centro Interdipartimentale di Ricerca sull'Ipertensione Arteriosa e le Patologie Associate (CIRIAPA), Dipartimento di Scienze Biomediche Avanzate, Università Federico II, Napoli, Italy
| |
Collapse
|
46
|
Garay-Gutiérrez NF, Hernandez-Fuentes CP, García-Rivas G, Lavandero S, Guerrero-Beltrán CE. Vaccines against components of the renin-angiotensin system. Heart Fail Rev 2020; 26:711-726. [PMID: 32995973 PMCID: PMC7524378 DOI: 10.1007/s10741-020-10033-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/21/2020] [Indexed: 12/26/2022]
Abstract
Even though effective drugs for treating hypertension are available, a great percentage of patients have inadequate control of their blood pressure. Unwanted side effects and inappropriate oral drug adherence are important factors that contribute to the global problem of uncontrolled hypertension. Vaccination could provide a revolutionary therapy with long-lasting effects, increasing patient compliance and therefore better control of high blood pressure. Nowadays, current immunization approaches against hypertension target renin, angiotensin I, angiotensin II, and angiotensin II type 1 receptor, key elements of the renin–angiotensin system. This article reviews the different vaccination attempts with proteins and peptides against the different molecules of the renin–angiotensin system in the last two decades, safety issues, and other novel prospects biomarkers in hypertension, and summarizes the potential of this immunomodulatory approach in clinical practice.
Collapse
Affiliation(s)
- Noé Francisco Garay-Gutiérrez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Medicina Cardiovascular y Metabolómica, Tecnologico de Monterrey, Monterrey, NL, Mexico
| | - Carolina Paz Hernandez-Fuentes
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Gerardo García-Rivas
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Medicina Cardiovascular y Metabolómica, Tecnologico de Monterrey, Monterrey, NL, Mexico.,Hospital Zambrano Hellion, TecSalud, Centro de Investigación Biomédica, Tecnologico de Monterrey, San Pedro Garza García, NL, Mexico
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Department of Internal Medicine, Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Carlos Enrique Guerrero-Beltrán
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Medicina Cardiovascular y Metabolómica, Tecnologico de Monterrey, Monterrey, NL, Mexico. .,Hospital Zambrano Hellion, TecSalud, Centro de Investigación Biomédica, Tecnologico de Monterrey, San Pedro Garza García, NL, Mexico.
| |
Collapse
|
47
|
Townsend A. Autoimmunity to ACE2 as a possible cause of tissue inflammation in Covid-19. Med Hypotheses 2020; 144:110043. [PMID: 32795830 PMCID: PMC7382330 DOI: 10.1016/j.mehy.2020.110043] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 06/23/2020] [Indexed: 11/18/2022]
Abstract
Hypothesis The delayed lung damage after SARS-CoV-2 infection may be caused by an autoimmune response to ACE2 induced by forced presentation of the ACE2 protein in a complex with CoV Spike in Fc Receptor positive Antigen Presenting Cells in the lung. The likelihood that this hypothesis is valid is low, but it is easily tested. Testable predictions 1) Autoantibodies and T cells to ACE2 may be found in patients with the lung damage but not in those without 2) There may be an HLA linkage with the delayed lung disease 3) Vaccines based on the spike protein might initiate the process by amplifying Fc mediated uptake of ACE2-Spike complexes into APCs. Practical implications The development of autoantibodies to ACE2 might predict the development of the inflammatory phase of Covid-19 disease. It might be wise to consider engineering versions of the spike that no longer bind to ACE2 for inclusion in vaccines.
Collapse
Affiliation(s)
- Alain Townsend
- Human Immunology Unit, Weatherall Institute, Oxford OX39DS, United Kingdom.
| |
Collapse
|
48
|
Sandoval J, Del Valle-Mondragón L, Masso F, Zayas N, Pulido T, Teijeiro R, Gonzalez-Pacheco H, Olmedo-Ocampo R, Sisniega C, Paez-Arenas A, Pastelin-Hernandez G, Gomez-Arroyo J, Voelkel NF. Angiotensin converting enzyme 2 and angiotensin (1-7) axis in pulmonary arterial hypertension. Eur Respir J 2020; 56:13993003.02416-2019. [PMID: 32241831 DOI: 10.1183/13993003.02416-2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 03/21/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND In animal models of pulmonary arterial hypertension (PAH), angiotensin-converting enzyme (ACE)2 and angiotensin (Ang)-(1-7) have been shown to have vasodilatory, antiproliferative, antifibrotic and antihypertrophic properties. However, the status and role of the ACE2-Ang(1-7) axis in human PAH is incompletely understood. METHODS We studied 85 patients with a diagnosis of PAH of distinct aetiologies. 55 healthy blood donors paired for age and sex served as controls. Blood samples were obtained from the pulmonary artery in patients with PAH during right heart catheterisation. Peripheral blood was obtained for both groups. Ang(1-7) and -II were measured using zone capillary electrophoresis. Aldosterone, Ang(1-9), AngA and ACE2 were measured using ELISA, and ACE2 activity was determined enzymatically. RESULTS Of the 85 patients, 47 had idiopathic PAH, 25 had PAH associated with congenital heart disease and 13 had PAH associated with collagen vascular disease. Compared to controls, patients with PAH had a higher concentration of AngII (median 1.03, interquartile range 0.72-1.88 pmol·mL-1 versus 0.19, 0.10-0.37 pmol·mL-1; p<0.001) and of aldosterone (88.7, 58.7-132 ng·dL-1 versus 12.9, 9.55-19.9 ng·dL-1; p<0.001). Conversely, PAH patients had a lower concentration of Ang(1-7) than controls (0.69, 0.474-0.91 pmol·mL-1 versus 4.07, 2.82-6.73 pmol·mL-1; p<0.001), and a lower concentration of Ang(1-9) and AngA. Similarly, the ACE2 concentration was higher than in controls (8.7, 5.35-13.2 ng·mL-1 versus 4.53, 1.47-14.3 ng·mL-1; p=0.011), whereas the ACE2 activity was significantly reduced (1.88, 1.08-2.81 nmol·mL-1 versus 5.97, 3.1-17.8 nmol·mL-1; p<0.001). No significant differences were found among the three different aetiological forms of PAH. CONCLUSIONS The AngII-ACE2-Ang(1-7) axis appears to be altered in human PAH and we propose that this imbalance, in favour of AngII, plays a role in the pathogenesis of the severe PAH. Further mechanistic studies are warranted.
Collapse
Affiliation(s)
- Julio Sandoval
- Cardiopulmonary Dept, Instituto Nacional de Cardiologia, Mexico City, Mexico
| | | | - Felipe Masso
- Physiology and Molecular Biology Dept of the "Ignacio Chávez", National Institute of Cardiology, Mexico City, Mexico
| | - Nayeli Zayas
- Cardiopulmonary Dept, Instituto Nacional de Cardiologia, Mexico City, Mexico
| | - Tomás Pulido
- Cardiopulmonary Dept, Instituto Nacional de Cardiologia, Mexico City, Mexico
| | - Ricardo Teijeiro
- Cardiopulmonary Dept, Instituto Nacional de Cardiologia, Mexico City, Mexico
| | | | | | - Carlos Sisniega
- Cardiopulmonary Dept, Instituto Nacional de Cardiologia, Mexico City, Mexico
| | - Araceli Paez-Arenas
- Physiology and Molecular Biology Dept of the "Ignacio Chávez", National Institute of Cardiology, Mexico City, Mexico
| | | | - Jose Gomez-Arroyo
- Cardiopulmonary Dept, Instituto Nacional de Cardiologia, Mexico City, Mexico.,Division of Pulmonary and Critical Care Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Norbert F Voelkel
- Dept of Pulmonary Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
49
|
Michaud V, Deodhar M, Arwood M, Al Rihani SB, Dow P, Turgeon J. ACE2 as a Therapeutic Target for COVID-19; its Role in Infectious Processes and Regulation by Modulators of the RAAS System. J Clin Med 2020; 9:E2096. [PMID: 32635289 PMCID: PMC7408699 DOI: 10.3390/jcm9072096] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 06/25/2020] [Accepted: 07/02/2020] [Indexed: 01/08/2023] Open
Abstract
Angiotensin converting enzyme 2 (ACE2) is the recognized host cell receptor responsiblefor mediating infection by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). ACE2bound to tissue facilitates infectivity of SARS-CoV-2; thus, one could argue that decreasing ACE2tissue expression would be beneficial. However, ACE2 catalytic activity towards angiotensin I (AngI) and II (Ang II) mitigates deleterious effects associated with activation of the renin-angiotensinaldosteronesystem (RAAS) on several organs, including a pro-inflammatory status. At the tissuelevel, SARS-CoV-2 (a) binds to ACE2, leading to its internalization, and (b) favors ACE2 cleavage toform soluble ACE2: these actions result in decreased ACE2 tissue levels. Preserving tissue ACE2activity while preventing ACE2 shredding is expected to circumvent unrestrained inflammatoryresponse. Concerns have been raised around RAAS modulators and their effects on ACE2expression or catalytic activity. Various cellular and animal models report conflicting results invarious tissues. However, recent data from observational and meta-analysis studies in SARS-CoV-2-infected patients have concluded that RAAS modulators do not increase plasma ACE2 levels orsusceptibility to infection and are not associated with more severe diseases. This review presentsour current but evolving knowledge of the complex interplay between SARS-CoV-2 infection, ACE2levels, modulators of RAAS activity and the effects of RAAS modulators on ACE2 expression.
Collapse
Affiliation(s)
- Veronique Michaud
- Tabula Rasa HealthCare Precision Pharmacotherapy Research & Development Institute, Orlando, FL 32827, USA; (V.M.); (M.D.); (M.A.); (S.B.A.R.); (P.D.)
- Faculty of Pharmacy, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Malavika Deodhar
- Tabula Rasa HealthCare Precision Pharmacotherapy Research & Development Institute, Orlando, FL 32827, USA; (V.M.); (M.D.); (M.A.); (S.B.A.R.); (P.D.)
| | - Meghan Arwood
- Tabula Rasa HealthCare Precision Pharmacotherapy Research & Development Institute, Orlando, FL 32827, USA; (V.M.); (M.D.); (M.A.); (S.B.A.R.); (P.D.)
| | - Sweilem B Al Rihani
- Tabula Rasa HealthCare Precision Pharmacotherapy Research & Development Institute, Orlando, FL 32827, USA; (V.M.); (M.D.); (M.A.); (S.B.A.R.); (P.D.)
| | - Pamela Dow
- Tabula Rasa HealthCare Precision Pharmacotherapy Research & Development Institute, Orlando, FL 32827, USA; (V.M.); (M.D.); (M.A.); (S.B.A.R.); (P.D.)
| | - Jacques Turgeon
- Tabula Rasa HealthCare Precision Pharmacotherapy Research & Development Institute, Orlando, FL 32827, USA; (V.M.); (M.D.); (M.A.); (S.B.A.R.); (P.D.)
- Faculty of Pharmacy, Université de Montréal, Montreal, QC H3C 3J7, Canada
| |
Collapse
|
50
|
Bourgonje AR, Abdulle AE, Timens W, Hillebrands JL, Navis GJ, Gordijn SJ, Bolling MC, Dijkstra G, Voors AA, Osterhaus AD, van der Voort PH, Mulder DJ, van Goor H. Angiotensin-converting enzyme 2 (ACE2), SARS-CoV-2 and the pathophysiology of coronavirus disease 2019 (COVID-19). J Pathol 2020; 251:228-248. [PMID: 32418199 PMCID: PMC7276767 DOI: 10.1002/path.5471] [Citation(s) in RCA: 687] [Impact Index Per Article: 171.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023]
Abstract
Angiotensin-converting enzyme 2 (ACE2) has been established as the functional host receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus responsible for the current devastating worldwide pandemic of coronavirus disease 2019 (COVID-19). ACE2 is abundantly expressed in a variety of cells residing in many different human organs. In human physiology, ACE2 is a pivotal counter-regulatory enzyme to ACE by the breakdown of angiotensin II, the central player in the renin-angiotensin-aldosterone system (RAAS) and the main substrate of ACE2. Many factors have been associated with both altered ACE2 expression and COVID-19 severity and progression, including age, sex, ethnicity, medication, and several co-morbidities, such as cardiovascular disease and metabolic syndrome. Although ACE2 is widely distributed in various human tissues and many of its determinants have been well recognised, ACE2-expressing organs do not equally participate in COVID-19 pathophysiology, implying that other mechanisms are involved in orchestrating cellular infection resulting in tissue damage. Reports of pathologic findings in tissue specimens of COVID-19 patients are rapidly emerging and confirm the established role of ACE2 expression and activity in disease pathogenesis. Identifying pathologic changes caused by SARS-CoV-2 infection is crucially important as it has major implications for understanding COVID-19 pathophysiology and the development of evidence-based treatment strategies. Currently, many interventional strategies are being explored in ongoing clinical trials, encompassing many drug classes and strategies, including antiviral drugs, biological response modifiers, and RAAS inhibitors. Ultimately, prevention is key to combat COVID-19 and appropriate measures are being taken accordingly, including development of effective vaccines. In this review, we describe the role of ACE2 in COVID-19 pathophysiology, including factors influencing ACE2 expression and activity in relation to COVID-19 severity. In addition, we discuss the relevant pathological changes resulting from SARS-CoV-2 infection. Finally, we highlight a selection of potential treatment modalities for COVID-19. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Arno R Bourgonje
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Amaal E Abdulle
- Department of Internal Medicine, Division of Vascular Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Wim Timens
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gerjan J Navis
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sanne J Gordijn
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marieke C Bolling
- Department of Dermatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gerard Dijkstra
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Adriaan A Voors
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Albert Dme Osterhaus
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hannover, Germany
| | - Peter Hj van der Voort
- Department of Critical Care Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Douwe J Mulder
- Department of Internal Medicine, Division of Vascular Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|