1
|
Tsiftsoglou SA, Gavriilaki E. A potential bimodal interplay between heme and complement factor H 402H in the deregulation of the complement alternative pathway by SARS-CoV-2. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2024; 126:105698. [PMID: 39643072 DOI: 10.1016/j.meegid.2024.105698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 11/29/2024] [Accepted: 12/01/2024] [Indexed: 12/09/2024]
Abstract
The recent discovery that the trimeric SARS-CoV-2 spike S glycoprotein carries heme within an NTD domain pocket of the S1 subunits, suggested that this virus may be cleverly utilizing heme, in addition to the S1 RBD domains, for invading target cells carrying a specific entry receptor like ACE2, TMEM106B and others. Studies during the COVID-19 pandemic revealed that the infectivity of this virus depends on cell surface heparan sulfate and that the infection induces non-canonical activation of the Complement Alternative pathway (AP) on the surface of infected cells. In our recent COVID-19 genomic studies, among the coding SNPs of interest we also detected the presence of the CFH rs1061170, rs800292 and rs1065489 within all the infected patient subgroups examined. The minor C allele of rs1061170 encodes CFH 402H that over the years has been associated with diseases characterized by complement dysregulation namely the age-related macular degeneration (AMD) and the atypical haemolytic uremic syndrome (aHUS). Also, more recently with the diminishment of CD4+ T cell responses with ageing. The rs800292 minor allele A encodes CFH 62I that supports enhanced cofactor activity for Complement factor I (CFI). Also, the rs1065489 minor allele T encodes CFH 936D and is located within the CCP16 domain that influences the affinity of CFH with extracellular laminins. A subsequent computational analysis revealed that the CFH residue 402 is located centrally within a heme-binding motif (HBM) in domain CCP7 (398YNQNYGRKF406). Heme on the viral spike glycoprotein S1 subunit could recruit CFH 402H for masking free viral particles from opsonisation, and when in proximity to cell surface, act as a bait disrupting CFH 402H from the heparan sulphate coat of the target cells. Publicly available genetic data for European populations indicate that the minor C allele of rs1061170 is present only in haplotypes that carry the major alleles of rs800292 and rs1065489. This combination encodes for CFH 402H that exhibits increased biochemical affinity for heme in proximity, without enhanced cofactor activity for CFI and weaker association with the extracellular matrix. In the theatre of infection, this combination can promote heme-mediated viral infection with weaker complement opsonisation and potential AP deregulation. This strategy may be evolutionary conserved among various classes of infectious agents.
Collapse
Affiliation(s)
- Stefanos A Tsiftsoglou
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece; Department of Biomedical Sciences, School of Health Sciences, Alexander Campus, International Hellenic University, Sindos, 57400, Greece.
| | - Eleni Gavriilaki
- 2(nd) Propedeutic Department of Internal Medicine, Hippocration General Hospital, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| |
Collapse
|
2
|
Gebetsberger L, Malekshahi Z, Teutsch A, Tajti G, Fontaine F, Marella N, Mueller A, Prantl L, Stockinger H, Stoiber H, Ohradanova-Repic A. SARS-CoV-2 hijacks host CD55, CD59 and factor H to impair antibody-dependent complement-mediated lysis. Emerg Microbes Infect 2024; 13:2417868. [PMID: 39435487 PMCID: PMC11520101 DOI: 10.1080/22221751.2024.2417868] [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: 05/26/2024] [Revised: 09/18/2024] [Accepted: 10/13/2024] [Indexed: 10/23/2024]
Abstract
The complement system is a vital anti-microbial defence mechanism against circulating pathogens. Excessive complement activation can have deleterious outcomes for the host and is consequently tightly modulated by a set of membrane-associated and fluid-phase regulators of complement activation (RCAs). Here, we demonstrate that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) hijacks host cellular RCA members CD55 and CD59 and serum-derived Factor H (FH) to resist antibody-dependent complement-mediated lysis triggered by immunized human sera. Blockage of the biological functions of virion-associated CD55 and CD59 and competition of FH recruitment with functionally inactive recombinant FH-derived short consensus repeats SCR18-20 restore SARS-CoV-2 complement sensitivity in a synergistic manner. Moreover, complement-mediated virolysis is dependent on classical pathway activation and does not occur in the absence of virus-specific antibodies. Altogether, our findings present an intriguing immune escape mechanism that provides novel insights into the immunopathology observed in severe coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Laura Gebetsberger
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Zahra Malekshahi
- Medical University of Innsbruck, Institute of Virology, Innsbruck, Austria
| | - Aron Teutsch
- Medical University of Innsbruck, Institute of Virology, Innsbruck, Austria
| | - Gabor Tajti
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Frédéric Fontaine
- CeMM – Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Nara Marella
- CeMM – Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - André Mueller
- CeMM – Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Lena Prantl
- Medical University of Innsbruck, Institute of Virology, Innsbruck, Austria
| | - Hannes Stockinger
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Heribert Stoiber
- Medical University of Innsbruck, Institute of Virology, Innsbruck, Austria
| | - Anna Ohradanova-Repic
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| |
Collapse
|
3
|
Wang G, Yang L, Xu X, Guo W, Sun L, Wang Y, Cheng W, Ye N, Kong L, Zhao X, Cheng H. SARS-CoV-2 Protein Deposition Enhances Renal Complement Activation and Aggravates Kidney Injury in Membranous Nephropathy After COVID-19. Kidney Int Rep 2024; 9:3145-3155. [PMID: 39534192 PMCID: PMC11551107 DOI: 10.1016/j.ekir.2024.08.006] [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: 02/02/2024] [Revised: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction COVID-19 has been reported to be associated with the occurrence and recurrence of membranous nephropathy (MN). The clinicopathological characteristics and complement system activation of MN after COVID-19 are unclear. Methods A total of 38 patients with biopsy-proven MN who developed new-onset proteinuria after COVID-19 were enrolled in this study. One hundred patients with primary MN diagnosed before the COVID-19 pandemic were the control. Renal immunohistochemical staining for SARS-CoV-2 nucleocapsid protein was performed in 38 patients with MN after COVID-19. Serum membrane attack complex (MAC) was detected by enzyme-linked immunosorbent assay. Glomerular staining for the complement proteins in different pathways were detected by immunohistochemistry. Results Thirteen of 38 patients had positive staining for SARS-CoV-2 nucleocapsid protein. Compared with the control patients, the clinical manifestations were more severe in patients after COVID-19. Patients with positive SARS-CoV-2 staining had a higher proportion of nephrotic syndrome, lower level of serum albumin, and greater severity of renal interstitial fibrosis than those of patients with negative SARS-CoV-2 staining. Serum MAC level and renal MAC staining intensity of MN after COVID-19 were significantly higher than those of the control patients. MAC expression in MN patients with positive SARS-CoV-2 staining was stronger than that in both control patients and MN after COVID-19 with negative SARS-CoV-2 staining. The expression trend of factor H was consistent with that of MAC. Conclusion Excessive activation of the complement system aggravated symptoms in MN after COVID-19. Therapeutic strategy targeting the complement system may need to be considered.
Collapse
Affiliation(s)
- Guoqin Wang
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Lei Yang
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xiaoyi Xu
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Weiyi Guo
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Lijun Sun
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yanyan Wang
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Wenrong Cheng
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Nan Ye
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Lingqiang Kong
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xiaoyi Zhao
- Division of Nephrology, Affiliated Hospital of Chifeng University, Neimenggu, China
| | - Hong Cheng
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
Avdonin PP, Blinova MS, Serkova AA, Komleva LA, Avdonin PV. Immunity and Coagulation in COVID-19. Int J Mol Sci 2024; 25:11267. [PMID: 39457048 PMCID: PMC11508857 DOI: 10.3390/ijms252011267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/23/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Discovered in late 2019, the SARS-CoV-2 coronavirus has caused the largest pandemic of the 21st century, claiming more than seven million lives. In most cases, the COVID-19 disease caused by the SARS-CoV-2 virus is relatively mild and affects only the upper respiratory tract; it most often manifests itself with fever, chills, cough, and sore throat, but also has less-common mild symptoms. In most cases, patients do not require hospitalization, and fully recover. However, in some cases, infection with the SARS-CoV-2 virus leads to the development of a severe form of COVID-19, which is characterized by the development of life-threatening complications affecting not only the lungs, but also other organs and systems. In particular, various forms of thrombotic complications are common among patients with a severe form of COVID-19. The mechanisms for the development of thrombotic complications in COVID-19 remain unclear. Accumulated data indicate that the pathogenesis of severe COVID-19 is based on disruptions in the functioning of various innate immune systems. The key role in the primary response to a viral infection is assigned to two systems. These are the pattern recognition receptors, primarily members of the toll-like receptor (TLR) family, and the complement system. Both systems are the first to engage in the fight against the virus and launch a whole range of mechanisms aimed at its rapid elimination. Normally, their joint activity leads to the destruction of the pathogen and recovery. However, disruptions in the functioning of these innate immune systems in COVID-19 can cause the development of an excessive inflammatory response that is dangerous for the body. In turn, excessive inflammation entails activation of and damage to the vascular endothelium, as well as the development of the hypercoagulable state observed in patients seriously ill with COVID-19. Activation of the endothelium and hypercoagulation lead to the development of thrombosis and, as a result, damage to organs and tissues. Immune-mediated thrombotic complications are termed "immunothrombosis". In this review, we discuss in detail the features of immunothrombosis associated with SARS-CoV-2 infection and its potential underlying mechanisms.
Collapse
Affiliation(s)
| | | | | | | | - Pavel V. Avdonin
- Koltzov Institute of Developmental Biology RAS, ul. Vavilova, 26, 119334 Moscow, Russia; (P.P.A.)
| |
Collapse
|
5
|
Szachowicz PJ, Wohlford-Lenane C, Heinen CJ, Ghimire S, Xue B, Boly TJ, Verma A, MašinoviĆ L, Bermick JR, Perlman S, Meyerholz DK, Pezzulo AA, Zhang Y, Smith RJ, McCray PB. A predominately pulmonary activation of complement in a mouse model of severe COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.31.596892. [PMID: 38895461 PMCID: PMC11185570 DOI: 10.1101/2024.05.31.596892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Evidence from in vitro studies and observational human disease data suggest the complement system plays a significant role in SARS-CoV-2 pathogenesis, although how complement dysregulation develops in patients with severe COVID-19 is unknown. Here, using a mouse-adapted SARS-CoV-2 virus (SARS2-N501YMA30) and a mouse model of severe COVID-19, we identify significant serologic and pulmonary complement activation following infection. We observed C3 activation in airway and alveolar epithelia, and in pulmonary vascular endothelia. Our evidence suggests that while the alternative pathway is the primary route of complement activation, components of both the alternative and classical pathways are produced locally by respiratory epithelial cells following infection, and increased in primary cultures of human airway epithelia in response to cytokine exposure. This locally generated complement response appears to precede and subsequently drive lung injury and inflammation. Results from this mouse model recapitulate findings in humans, which suggest sex-specific variance in complement activation, with predilection for increased C3 activity in males, a finding that may correlate with more severe disease. Our findings indicate that complement activation is a defining feature of severe COVID-19 in mice and lay the foundation for further investigation into the role of complement in COVID-19.
Collapse
Affiliation(s)
- Peter J. Szachowicz
- Department of Internal Medicine, The University of Iowa, Division of Pulmonary, Critical Care, and Occupational Medicine, Iowa City, IA, 52242
| | | | - Cobey J. Heinen
- Molecular Otolaryngology and Renal Research Laboratories, Carver College of Medicine, University of Iowa, USA
| | - Shreya Ghimire
- Department of Internal Medicine, The University of Iowa, Division of Pulmonary, Critical Care, and Occupational Medicine, Iowa City, IA, 52242
| | - Biyun Xue
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA, 52242
| | - Timothy J. Boly
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA, 52242
| | - Abhishek Verma
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, 52242
| | - Leila MašinoviĆ
- Department of Internal Medicine, The University of Iowa, Division of Pulmonary, Critical Care, and Occupational Medicine, Iowa City, IA, 52242
| | - Jennifer R. Bermick
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA, 52242
| | - Stanley Perlman
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA, 52242
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, 52242
| | | | - Alejandro A. Pezzulo
- Department of Internal Medicine, The University of Iowa, Division of Pulmonary, Critical Care, and Occupational Medicine, Iowa City, IA, 52242
| | - Yuzhou Zhang
- Molecular Otolaryngology and Renal Research Laboratories, Carver College of Medicine, University of Iowa, USA
| | - Richard J.H. Smith
- Molecular Otolaryngology and Renal Research Laboratories, Carver College of Medicine, University of Iowa, USA
| | - Paul B. McCray
- Stead Family Department of Pediatrics, The University of Iowa, Iowa City, IA, 52242
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, 52242
| |
Collapse
|
6
|
Guo WY, Wang GQ, Kong LQ, Sun LJ, Xu XY, Cheng WR, Dong HR, Cheng H. Complement system is overactivated in patients with IgA nephropathy after COVID-19. Clin Immunol 2024; 263:110232. [PMID: 38701960 DOI: 10.1016/j.clim.2024.110232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/26/2024] [Indexed: 05/06/2024]
Abstract
IgA nephropathy (IgAN), which has been confirmed as a complement mediated autoimmune disease, is also one form of glomerulonephritis associated with COVID-19. Here, we aim to investigate the clinical and immunological characteristics of patients with IgAN after COVID-19. The level of plasma level of C5a (p < 0.001), soluble C5b-9 (p = 0.018), FHR5 (p < 0.001) were all significantly higher in Group CoV (33 patients with renal biopsy-proven IgAN experienced COVID-19) compared with Group non-CoV (44 patients with IgAN without COVID-19), respectively. Compared with Group non-CoV, the intensity of glomerular C4d (p = 0.017) and MAC deposition (p < 0.001) and Gd-IgA1 deposition (p = 0.005) were much stronger in Group CoV. Our finding revealed that for IgAN after COVID-19, mucosal immune responses to SARS-CoV-2 infection may result in the overactivation of systemic and renal local complement system, and increased glomerular deposition of Gd-IgA1, which may lead to renal dysfunction and promote renal progression in IgAN patients.
Collapse
Affiliation(s)
- Wei-Yi Guo
- Renal Division, Department of Medicine, Beijing Anzhen Hospital, Capital Medical University, China
| | - Guo-Qin Wang
- Renal Division, Department of Medicine, Beijing Anzhen Hospital, Capital Medical University, China
| | - Ling-Qiang Kong
- Renal Division, Department of Medicine, Beijing Anzhen Hospital, Capital Medical University, China
| | - Li-Jun Sun
- Renal Division, Department of Medicine, Beijing Anzhen Hospital, Capital Medical University, China
| | - Xiao-Yi Xu
- Renal Division, Department of Medicine, Beijing Anzhen Hospital, Capital Medical University, China
| | - Wen-Rong Cheng
- Renal Division, Department of Medicine, Beijing Anzhen Hospital, Capital Medical University, China
| | - Hong-Rui Dong
- Renal Division, Department of Medicine, Beijing Anzhen Hospital, Capital Medical University, China
| | - Hong Cheng
- Renal Division, Department of Medicine, Beijing Anzhen Hospital, Capital Medical University, China.
| |
Collapse
|
7
|
Bermejo-Jambrina M, van der Donk LE, van Hamme JL, Wilflingseder D, de Bree G, Prins M, de Jong M, Nieuwkerk P, van Gils MJ, Kootstra NA, Geijtenbeek TB. Control of complement-induced inflammatory responses to SARS-CoV-2 infection by anti-SARS-CoV-2 antibodies. EMBO J 2024; 43:1135-1163. [PMID: 38418557 PMCID: PMC10987522 DOI: 10.1038/s44318-024-00061-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 01/31/2024] [Accepted: 02/06/2024] [Indexed: 03/01/2024] Open
Abstract
Dysregulated immune responses contribute to the excessive and uncontrolled inflammation observed in severe COVID-19. However, how immunity to SARS-CoV-2 is induced and regulated remains unclear. Here, we uncover the role of the complement system in the induction of innate and adaptive immunity to SARS-CoV-2. Complement rapidly opsonizes SARS-CoV-2 particles via the lectin pathway. Complement-opsonized SARS-CoV-2 efficiently induces type-I interferon and pro-inflammatory cytokine responses via activation of dendritic cells, which are inhibited by antibodies against the complement receptors (CR) 3 and 4. Serum from COVID-19 patients, or monoclonal antibodies against SARS-CoV-2, attenuate innate and adaptive immunity induced by complement-opsonized SARS-CoV-2. Blocking of CD32, the FcγRII antibody receptor of dendritic cells, restores complement-induced immunity. These results suggest that opsonization of SARS-CoV-2 by complement is involved in the induction of innate and adaptive immunity to SARS-CoV-2 in the acute phase of infection. Subsequent antibody responses limit inflammation and restore immune homeostasis. These findings suggest that dysregulation of the complement system and FcγRII signaling may contribute to severe COVID-19.
Collapse
Affiliation(s)
- Marta Bermejo-Jambrina
- Department of Experimental Immunology, Amsterdam UMC location AMC, Amsterdam, The Netherlands.
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands.
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria.
| | - Lieve Eh van der Donk
- Department of Experimental Immunology, Amsterdam UMC location AMC, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands
| | - John L van Hamme
- Department of Experimental Immunology, Amsterdam UMC location AMC, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands
| | - Doris Wilflingseder
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Godelieve de Bree
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands
- Department of Internal Medicine, Amsterdam UMC location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Maria Prins
- Department of Internal Medicine, Amsterdam UMC location AMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Infectious Diseases, Public Health Service of Amsterdam, GGD, Amsterdam, The Netherlands
| | - Menno de Jong
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC location AMC University of Amsterdam, Amsterdam, The Netherlands
| | - Pythia Nieuwkerk
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands
- Department of Infectious Diseases, Public Health Service of Amsterdam, GGD, Amsterdam, The Netherlands
- Department of Medical Psychology (J3-2019-1), Amsterdam UMC location AMC University of Amsterdam, Amsterdam, The Netherlands
| | - Marit J van Gils
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC location AMC University of Amsterdam, Amsterdam, The Netherlands
| | - Neeltje A Kootstra
- Department of Experimental Immunology, Amsterdam UMC location AMC, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands
| | - Teunis Bh Geijtenbeek
- Department of Experimental Immunology, Amsterdam UMC location AMC, Amsterdam, The Netherlands.
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands.
| |
Collapse
|
8
|
Van Damme KFA, Hoste L, Declercq J, De Leeuw E, Maes B, Martens L, Colman R, Browaeys R, Bosteels C, Verwaerde S, Vermeulen N, Lameire S, Debeuf N, Deckers J, Stordeur P, Depuydt P, Van Braeckel E, Vandekerckhove L, Guilliams M, Schetters STT, Haerynck F, Tavernier SJ, Lambrecht BN. A complement atlas identifies interleukin-6-dependent alternative pathway dysregulation as a key druggable feature of COVID-19. Sci Transl Med 2023; 15:eadi0252. [PMID: 37611083 DOI: 10.1126/scitranslmed.adi0252] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 08/02/2023] [Indexed: 08/25/2023]
Abstract
Improvements in COVID-19 treatments, especially for the critically ill, require deeper understanding of the mechanisms driving disease pathology. The complement system is not only a crucial component of innate host defense but can also contribute to tissue injury. Although all complement pathways have been implicated in COVID-19 pathogenesis, the upstream drivers and downstream effects on tissue injury remain poorly defined. We demonstrate that complement activation is primarily mediated by the alternative pathway, and we provide a comprehensive atlas of the complement alterations around the time of respiratory deterioration. Proteomic and single-cell sequencing mapping across cell types and tissues reveals a division of labor between lung epithelial, stromal, and myeloid cells in complement production, in addition to liver-derived factors. We identify IL-6 and STAT1/3 signaling as an upstream driver of complement responses, linking complement dysregulation to approved COVID-19 therapies. Furthermore, an exploratory proteomic study indicates that inhibition of complement C5 decreases epithelial damage and markers of disease severity. Collectively, these results support complement dysregulation as a key druggable feature of COVID-19.
Collapse
Affiliation(s)
- Karel F A Van Damme
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Levi Hoste
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University, Ghent, Belgium
| | - Jozefien Declercq
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Elisabeth De Leeuw
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Bastiaan Maes
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Liesbet Martens
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
- Laboratory of Myeloid Cell Biology in Tissue Damage and Inflammation, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Biomedical Molecular Biology, Faculty of Science, Ghent University, Belgium
| | - Roos Colman
- Biostatistics Unit, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Robin Browaeys
- Bioinformatics Expertise Unit, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Cédric Bosteels
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
- Respiratory Infection and Defense Lab, Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Stijn Verwaerde
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
| | - Nicky Vermeulen
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University, Ghent, Belgium
| | - Sahine Lameire
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
| | - Nincy Debeuf
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
| | - Julie Deckers
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
| | - Patrick Stordeur
- Belgian National Reference Center for the Complement System, Laboratory of Immunology, LHUB-ULB, Université Libre de Bruxelles, Brussels, Belgium
| | - Pieter Depuydt
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Intensive Care Unit, Ghent University Hospital, Ghent, Belgium
| | - Eva Van Braeckel
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
- Respiratory Infection and Defense Lab, Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Linos Vandekerckhove
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Biomedical Molecular Biology, Faculty of Science, Ghent University, Belgium
- HIV Cure Research Center, Department of Internal Medicine and Pediatrics, Ghent University and Ghent University Hospital, 9000 Ghent, Belgium
| | - Martin Guilliams
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
- Laboratory of Myeloid Cell Biology in Tissue Damage and Inflammation, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Biomedical Molecular Biology, Faculty of Science, Ghent University, Belgium
| | - Sjoerd T T Schetters
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
| | - Filomeen Haerynck
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University, Ghent, Belgium
| | - Simon J Tavernier
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University, Ghent, Belgium
| | - Bart N Lambrecht
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
| |
Collapse
|
9
|
Beirag N, Varghese PM, Neto MM, Al Aiyan A, Khan HA, Qablan M, Shamji MH, Sim RB, Temperton N, Kishore U. Complement Activation-Independent Attenuation of SARS-CoV-2 Infection by C1q and C4b-Binding Protein. Viruses 2023; 15:1269. [PMID: 37376569 DOI: 10.3390/v15061269] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
The complement system is a key component of the innate immune response to viruses and proinflammatory events. Exaggerated complement activation has been attributed to the induction of a cytokine storm in severe SARS-CoV-2 infection. However, there is also an argument for the protective role of complement proteins, given their local synthesis or activation at the site of viral infection. This study investigated the complement activation-independent role of C1q and C4b-binding protein (C4BP) against SARS-CoV-2 infection. The interactions of C1q, its recombinant globular heads, and C4BP with the SARS-CoV-2 spike and receptor binding domain (RBD) were examined using direct ELISA. In addition, RT-qPCR was used to evaluate the modulatory effect of these complement proteins on the SARS-CoV-2-mediated immune response. Cell binding and luciferase-based viral entry assays were utilised to assess the effects of C1q, its recombinant globular heads, and C4BP on SARS-CoV-2 cell entry. C1q and C4BP bound directly to SARS-CoV-2 pseudotype particles via the RBD domain of the spike protein. C1q via its globular heads and C4BP were found to reduce binding as well as viral transduction of SARS-CoV-2 spike protein expressing lentiviral pseudotypes into transfected A549 cells expressing human ACE2 and TMPRSS2. Furthermore, the treatment of the SARS-CoV-2 spike, envelope, nucleoprotein, and membrane protein expressing alphaviral pseudotypes with C1q, its recombinant globular heads, or C4BP triggered a reduction in mRNA levels of proinflammatory cytokines and chemokines such as IL-1β, IL-8, IL-6, TNF-α, IFN-α, and RANTES (as well as NF-κB) in A549 cells expressing human ACE2 and TMPRSS2. In addition, C1q and C4BP treatment also reduced SARS-CoV-2 pseudotype infection-mediated NF-κB activation in A549 cells expressing human ACE2 and TMPRSS2. C1q and C4BP are synthesised primarily by hepatocytes; however, they are also produced by macrophages, and alveolar type II cells, respectively, locally at the pulmonary site. These findings support the notion that the locally produced C1q and C4BP can be protective against SARS-CoV-2 infection in a complement activation-independent manner, offering immune resistance by inhibiting virus binding to target host cells and attenuating the infection-associated inflammatory response.
Collapse
Affiliation(s)
- Nazar Beirag
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK
| | - Praveen M Varghese
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| | - Martin Mayora Neto
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent and Greenwich, Kent ME4 4TB, UK
| | - Ahmad Al Aiyan
- Department of Veterinary Medicine, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Haseeb A Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh 4545, Saudi Arabia
| | - Moneeb Qablan
- Department of Veterinary Medicine, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Mohamed H Shamji
- Immunomodulation and Tolerance Group, Department of Allergy and Clinical Immunology, National Heart and Lung Institute, Imperial College London, London SW7 2BX, UK
| | - Robert B Sim
- MRC Immunochemistry Unit, Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent and Greenwich, Kent ME4 4TB, UK
| | - Uday Kishore
- Department of Veterinary Medicine, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|
10
|
Zelek WM, Harrison RA. Complement and COVID-19: Three years on, what we know, what we don't know, and what we ought to know. Immunobiology 2023; 228:152393. [PMID: 37187043 PMCID: PMC10174470 DOI: 10.1016/j.imbio.2023.152393] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 05/17/2023]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus was identified in China in 2019 as the causative agent of COVID-19, and quickly spread throughout the world, causing over 7 million deaths, of which 2 million occurred prior to the introduction of the first vaccine. In the following discussion, while recognising that complement is just one of many players in COVID-19, we focus on the relationship between complement and COVID-19 disease, with limited digression into directly-related areas such as the relationship between complement, kinin release, and coagulation. Prior to the 2019 COVID-19 outbreak, an important role for complement in coronavirus diseases had been established. Subsequently, multiple investigations of patients with COVID-19 confirmed that complement dysregulation is likely to be a major driver of disease pathology, in some, if not all, patients. These data fuelled evaluation of many complement-directed therapeutic agents in small patient cohorts, with claims of significant beneficial effect. As yet, these early results have not been reflected in larger clinical trials, posing questions such as who to treat, appropriate time to treat, duration of treatment, and optimal target for treatment. While significant control of the pandemic has been achieved through a global scientific and medical effort to comprehend the etiology of the disease, through extensive SARS-CoV-2 testing and quarantine measures, through vaccine development, and through improved therapy, possibly aided by attenuation of the dominant strains, it is not yet over. In this review, we summarise complement-relevant literature, emphasise its main conclusions, and formulate a hypothesis for complement involvement in COVID-19. Based on this we make suggestions as to how any future outbreak might be better managed in order to minimise impact on patients.
Collapse
Affiliation(s)
- Wioleta M Zelek
- Dementia Research Institute and Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | | |
Collapse
|
11
|
Nakayama EE, Shioda T. SARS-CoV-2 Related Antibody-Dependent Enhancement Phenomena In Vitro and In Vivo. Microorganisms 2023; 11:microorganisms11041015. [PMID: 37110438 PMCID: PMC10145615 DOI: 10.3390/microorganisms11041015] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/07/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Antibody-dependent enhancement (ADE) is a phenomenon in which antibodies produced in the body after infection or vaccination may enhance subsequent viral infections in vitro and in vivo. Although rare, symptoms of viral diseases are also enhanced by ADE following infection or vaccination in vivo. This is thought to be due to the production of antibodies with low neutralizing activity that bind to the virus and facilitate viral entry, or antigen-antibody complexes that cause airway inflammation, or a predominance of T-helper 2 cells among the immune system cells which leads to excessive eosinophilic tissue infiltration. Notably, ADE of infection and ADE of disease are different phenomena that overlap. In this article, we will describe the three types of ADE: (1) Fc receptor (FcR)-dependent ADE of infection in macrophages, (2) FcR-independent ADE of infection in other cells, and (3) FcR-dependent ADE of cytokine production in macrophages. We will describe their relationship to vaccination and natural infection, and discuss the possible involvement of ADE phenomena in COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Emi E Nakayama
- Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan
| | - Tatsuo Shioda
- Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan
| |
Collapse
|
12
|
Goff LM, Davies K, Zelek WM, Kodosaki E, Hakim O, Lockhart S, O’Rahilly S, Morgan BP. Ethnic differences in complement system biomarkers and their association with metabolic health in men of Black African and White European ethnicity. Clin Exp Immunol 2023; 212:52-60. [PMID: 36722378 PMCID: PMC10081104 DOI: 10.1093/cei/uxad011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 12/18/2022] [Accepted: 01/31/2023] [Indexed: 02/02/2023] Open
Abstract
Inflammation plays a fundamental role in the development of several metabolic diseases, including obesity and type 2 diabetes (T2D); the complement system has been implicated in their development. People of Black African (BA) ethnicity are disproportionately affected by T2D and other metabolic diseases but the impact of ethnicity on the complement system has not been explored. We investigated ethnic differences in complement biomarkers and activation status between men of BA and White European (WE) ethnicity and explored their association with parameters of metabolic health. We measured a panel of 15 complement components, regulators, and activation products in fasting plasma from 89 BA and 96 WE men. Ethnic differences were statistically validated. Association of complement biomarkers with metabolic health indices (BMI, waist circumference, insulin resistance, and HbA1c) were assessed in the groups. Plasma levels of the key complement components C3 and C4, the regulators clusterin and properdin and the activation marker iC3b were significantly higher in BA compared to WE men after age adjustment, while FD levels were significantly lower. C3 and C4 levels positively correlated with some or all markers of metabolic dysfunction in both ethnic groups while FD was inversely associated with HbA1c in both groups, and clusterin and properdin were inversely associated with some markers of metabolic dysfunction only in the WE group. Our findings of increased levels of complement components and activation products in BA compared to WE men suggest differences in complement regulation that may impact susceptibility to poor metabolic health.
Collapse
Affiliation(s)
- L M Goff
- Department of Nutritional Sciences, School of Population & Life Course Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, UK
| | - K Davies
- Dementia Research Institute Cardiff, School of Medicine, Cardiff University, Cardiff, UK
| | - W M Zelek
- Dementia Research Institute Cardiff, School of Medicine, Cardiff University, Cardiff, UK
| | - E Kodosaki
- Dementia Research Institute Cardiff, School of Medicine, Cardiff University, Cardiff, UK
| | - O Hakim
- Department of Nutritional Sciences, School of Population & Life Course Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, UK
- School of Life & Health Sciences, University of Roehampton, London, UK
| | - S Lockhart
- MRC Metabolic Diseases Unit & Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - S O’Rahilly
- MRC Metabolic Diseases Unit & Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - B P Morgan
- Dementia Research Institute Cardiff, School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
13
|
Tsiftsoglou SA, Gavriilaki E, Touloumenidou T, Koravou EE, Koutra M, Papayanni PG, Karali V, Papalexandri A, Varelas C, Chatzopoulou F, Chatzidimitriou M, Chatzidimitriou D, Veleni A, Rapti E, Kioumis I, Kaimakamis E, Bitzani M, Boumpas DT, Tsantes A, Sotiropoulos D, Papadopoulou A, Sakellari I, Kokoris S, Anagnostopoulos A. Targeted genotyping of COVID-19 patients reveals a signature of complement C3 and factor B coding SNPs associated with severe infection. Immunobiology 2023; 228:152351. [PMID: 36805858 PMCID: PMC9928680 DOI: 10.1016/j.imbio.2023.152351] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 01/19/2023] [Accepted: 02/11/2023] [Indexed: 02/17/2023]
Abstract
We have attempted to explore further the involvement of complement components in the host COVID-19 (Coronavirus disease-19) immune responses by targeted genotyping of COVID-19 adult patients and analysis for missense coding Single Nucleotide Polymorphisms (coding SNPs) of genes encoding Alternative pathway (AP) components. We have identified a small group of common coding SNPs in Survivors and Deceased individuals, present in either relatively similar frequencies (CFH and CFI SNPs) or with stark differences in their relative abundance (C3 and CFB SNPs). In addition, we have identified several sporadic, potentially protective, coding SNPs of C3, CFB, CFD, CFH, CFHR1 and CFI in Survivors. No coding SNPs were detected for CD46 and CD55. Our demographic analysis indicated that the C3 rs1047286 or rs2230199 coding SNPs were present in 60 % of all the Deceased patients (n = 25) (the rs2230199 in 67 % of all Deceased Males) and in 31 % of all the Survivors (n = 105, p = 0.012) (the rs2230199 in 25 % of all Survivor Males). When we analysed these two major study groups using the presence of the C3 rs1047286 or rs2230199 SNPs as potential biomarkers, we noticed the complete absence of the protective CFB rs12614 and rs641153 coding SNPs from Deceased Males compared to Females (p = 0.0023). We propose that in these individuals, C3 carrying the R102G and CFB lacking the R32W or the R32Q amino acid substitutions, may contribute to enhanced association dynamics of the C3bBb AP pre-convertase complex assembly, thus enabling the exploitation of the activation of the Complement Alternative pathway (AP) by SARS-CoV-2.
Collapse
Affiliation(s)
- Stefanos A Tsiftsoglou
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece.
| | - Eleni Gavriilaki
- Hematology Department-BMT Unit, G. Papanicolaou Hospital, Exochi, Thessaloniki 57010, Greece.
| | - Tasoula Touloumenidou
- Hematology Department-BMT Unit, G. Papanicolaou Hospital, Exochi, Thessaloniki 57010, Greece
| | | | - Maria Koutra
- Hematology Department-BMT Unit, G. Papanicolaou Hospital, Exochi, Thessaloniki 57010, Greece
| | | | - Vassiliki Karali
- Rheumatology and Clinical Immunology Unit, University General Hospital "Attikon", Αthens, Greece
| | - Apostolia Papalexandri
- Hematology Department-BMT Unit, G. Papanicolaou Hospital, Exochi, Thessaloniki 57010, Greece
| | - Christos Varelas
- Hematology Department-BMT Unit, G. Papanicolaou Hospital, Exochi, Thessaloniki 57010, Greece
| | - Fani Chatzopoulou
- Microbiology Department, Aristotle University of Thessaloniki, Greece
| | - Maria Chatzidimitriou
- Biomedical Sciences Alexander Campus International Hellenic University, Thessaloniki, Greece
| | | | - Anastasia Veleni
- Infectious Disease Committee, G Papanicolaou Hospital, Thessaloniki, Greece
| | - Evdoxia Rapti
- Laboratory of Hematology and Hospital Blood Transfusion Department, University General Hospital "Attikon", NKUA, Medical School, Athens, Greece
| | - Ioannis Kioumis
- Respiratory Failure Department, G Papanicolaou Hospital-Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Milly Bitzani
- 1st Intensive Care Unit, G Papanicolaou Hospital, Thessaloniki, Greece
| | - Dimitrios T Boumpas
- Rheumatology and Clinical Immunology Unit, University General Hospital "Attikon", Αthens, Greece
| | - Argyris Tsantes
- Laboratory of Hematology and Hospital Blood Transfusion Department, University General Hospital "Attikon", NKUA, Medical School, Athens, Greece
| | - Damianos Sotiropoulos
- Hematology Department-BMT Unit, G. Papanicolaou Hospital, Exochi, Thessaloniki 57010, Greece
| | - Anastasia Papadopoulou
- Hematology Department-BMT Unit, G. Papanicolaou Hospital, Exochi, Thessaloniki 57010, Greece
| | - Ioanna Sakellari
- Hematology Department-BMT Unit, G. Papanicolaou Hospital, Exochi, Thessaloniki 57010, Greece
| | - Styliani Kokoris
- Laboratory of Hematology and Hospital Blood Transfusion Department, University General Hospital "Attikon", NKUA, Medical School, Athens, Greece
| | | |
Collapse
|
14
|
Lim EHT, van Amstel RBE, de Boer VV, van Vught LA, de Bruin S, Brouwer MC, Vlaar APJ, van de Beek D. Complement activation in COVID-19 and targeted therapeutic options: A scoping review. Blood Rev 2023; 57:100995. [PMID: 35934552 PMCID: PMC9338830 DOI: 10.1016/j.blre.2022.100995] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/07/2022] [Accepted: 07/27/2022] [Indexed: 01/28/2023]
Abstract
Increasing evidence suggests that activation of the complement system plays a key role in the pathogenesis and disease severity of Coronavirus disease 2019 (COVID-19). We used a systematic approach to create an overview of complement activation in COVID-19 based on histopathological, preclinical, multiomics, observational and clinical interventional studies. A total of 1801 articles from PubMed, EMBASE and Cochrane was screened of which 157 articles were included in this scoping review. Histopathological, preclinical, multiomics and observational studies showed apparent complement activation through all three complement pathways and a correlation with disease severity and mortality. The complement system was targeted at different levels in COVID-19, of which C5 and C5a inhibition seem most promising. Adequately powered, double blind RCTs are necessary in order to further investigate the effect of targeting the complement system in COVID-19.
Collapse
Affiliation(s)
- Endry Hartono Taslim Lim
- Amsterdam UMC location University of Amsterdam, Department of Intensive Care Medicine, Meibergdreef 9, Amsterdam, the Netherlands,Amsterdam UMC Location University of Amsterdam, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam, the Netherlands,Amsterdam UMC location University of Amsterdam, Department of Neurology, Meibergdreef 9, Amsterdam, the Netherlands,Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Rombout Benjamin Ezra van Amstel
- Amsterdam UMC location University of Amsterdam, Department of Intensive Care Medicine, Meibergdreef 9, Amsterdam, the Netherlands,Amsterdam UMC Location University of Amsterdam, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam, the Netherlands
| | - Vieve Victoria de Boer
- Amsterdam UMC location University of Amsterdam, Department of Intensive Care Medicine, Meibergdreef 9, Amsterdam, the Netherlands
| | - Lonneke Alette van Vught
- Amsterdam UMC location University of Amsterdam, Department of Intensive Care Medicine, Meibergdreef 9, Amsterdam, the Netherlands,Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Amsterdam, the Netherlands
| | - Sanne de Bruin
- Amsterdam UMC location University of Amsterdam, Department of Intensive Care Medicine, Meibergdreef 9, Amsterdam, the Netherlands,Amsterdam UMC Location University of Amsterdam, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam, the Netherlands
| | - Matthijs Christian Brouwer
- Amsterdam UMC location University of Amsterdam, Department of Neurology, Meibergdreef 9, Amsterdam, the Netherlands,Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Alexander Petrus Johannes Vlaar
- Amsterdam UMC location University of Amsterdam, Department of Intensive Care Medicine, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam UMC Location University of Amsterdam, Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam, the Netherlands.
| | - Diederik van de Beek
- Amsterdam UMC location University of Amsterdam, Department of Neurology, Meibergdreef 9, Amsterdam, the Netherlands,Amsterdam Neuroscience, Amsterdam, the Netherlands
| |
Collapse
|
15
|
Tsai CL, Lai CC, Chen CY, Lee HS. The efficacy and safety of complement C5a inhibitors for patients with severe COVID-19: a systematic review and meta-analysis. Expert Rev Anti Infect Ther 2023; 21:77-86. [PMID: 36399686 DOI: 10.1080/14787210.2022.2150165] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND The clinical efficacy and safety of complement C5a inhibitors for patients with severe COVID-19 remains unclear. METHODS The PubMed, Embase, Cochrane Library, and ClinicalTrials.gov databases were searched from their inception to 27 September 2022. Only studies that assessed the usefulness of C5a inhibitors for the treatment of patients with severe COVID-19 patients were included. The primary outcome was the risk of 28-day mortality. RESULTS Six studies, including four randomized controlled trials (RCTs) and two non-RCTs, were included. The study group receiving C5a inhibitors had a significantly lower risk of mortality compared with the control group (23.6% [70/297] vs 39.2% [136/347]; odds ratio [OR], 0.53; 95% confidence interval [CI]: 0.37-0.76; P< 0.001), and no heterogeneity was detected (I2 = 0%; P= 0.58). Compared with control group, the study group was associated with a similar risk of serious adverse events (AEs) (OR, 0.84; 95% CI: 0.57-1.23; P0 = 0.37), infection (OR, 1.46; 95% CI: 0.77-2.79; P= 0.25) and acute kidney injury (OR, 0.89; 95% CI: 0.54-1.46; P= 0.64). CONCLUSION C5a inhibitors could help reduce the risk of mortality in patients with severe COVID-19 infection while being as safe as placebos. These findings support the promising role of C5a inhibitors in the treatment of severe COVID-19.
Collapse
Affiliation(s)
- Chi-Lun Tsai
- Department of Intensive Care Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Chih-Cheng Lai
- Division of Hospital Medicine, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan.,School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Ching-Yi Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, E-Da Hospital, Kaohsiung City, Taiwan.,School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Ho-Sheng Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, E-Da Hospital, Kaohsiung City, Taiwan.,School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| |
Collapse
|
16
|
Trivedi VS, Magnusen AF, Rani R, Marsili L, Slavotinek AM, Prows DR, Hopkin RJ, McKay MA, Pandey MK. Targeting the Complement-Sphingolipid System in COVID-19 and Gaucher Diseases: Evidence for a New Treatment Strategy. Int J Mol Sci 2022; 23:14340. [PMID: 36430817 PMCID: PMC9695449 DOI: 10.3390/ijms232214340] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/22/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2)-induced disease (COVID-19) and Gaucher disease (GD) exhibit upregulation of complement 5a (C5a) and its C5aR1 receptor, and excess synthesis of glycosphingolipids that lead to increased infiltration and activation of innate and adaptive immune cells, resulting in massive generation of pro-inflammatory cytokines, chemokines and growth factors. This C5a-C5aR1-glycosphingolipid pathway- induced pro-inflammatory environment causes the tissue damage in COVID-19 and GD. Strikingly, pharmaceutically targeting the C5a-C5aR1 axis or the glycosphingolipid synthesis pathway led to a reduction in glycosphingolipid synthesis and innate and adaptive immune inflammation, and protection from the tissue destruction in both COVID-19 and GD. These results reveal a common involvement of the complement and glycosphingolipid systems driving immune inflammation and tissue damage in COVID-19 and GD, respectively. It is therefore expected that combined targeting of the complement and sphingolipid pathways could ameliorate the tissue destruction, organ failure, and death in patients at high-risk of developing severe cases of COVID-19.
Collapse
Affiliation(s)
- Vyoma Snehal Trivedi
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
| | - Albert Frank Magnusen
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
| | - Reena Rani
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
| | - Luca Marsili
- Department of Neurology, James J. and Joan A. Gardner Center for Parkinson’s Disease and Movement Disorders, University of Cincinnati, 3113 Bellevue Ave, Cincinnati, OH 45219, USA
| | - Anne Michele Slavotinek
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, 3230 Eden Ave, Cincinnati, OH 45267, USA
| | - Daniel Ray Prows
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, 3230 Eden Ave, Cincinnati, OH 45267, USA
| | - Robert James Hopkin
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, 3230 Eden Ave, Cincinnati, OH 45267, USA
| | - Mary Ashley McKay
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
| | - Manoj Kumar Pandey
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, 3230 Eden Ave, Cincinnati, OH 45267, USA
| |
Collapse
|
17
|
Hurler L, Toonen EJM, Kajdácsi E, van Bree B, Brandwijk RJMGE, de Bruin W, Lyons PA, Bergamaschi L, Sinkovits G, Cervenak L, Würzner R, Prohászka Z. Distinction of early complement classical and lectin pathway activation via quantification of C1s/C1-INH and MASP-1/C1-INH complexes using novel ELISAs. Front Immunol 2022; 13:1039765. [PMID: 36420270 PMCID: PMC9677118 DOI: 10.3389/fimmu.2022.1039765] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/06/2022] [Indexed: 07/22/2023] Open
Abstract
The most commonly used markers to assess complement activation are split products that are produced through activation of all three pathways and are located downstream of C3. In contrast, C4d derives from the cleavage of C4 and indicates either classical (CP) or lectin pathway (LP) activation. Although C4d is perfectly able to distinguish between CP/LP and alternative pathway (AP) activation, no well-established markers are available to differentiate between early CP and LP activation. Active enzymes of both pathways (C1s/C1r for the CP, MASP-1/MASP-2 for the LP) are regulated by C1 esterase inhibitor (C1-INH) through the formation of covalent complexes. Aim of this study was to develop validated immunoassays detecting C1s/C1-INH and MASP-1/C1-INH complex levels. Measurement of the complexes reveals information about the involvement of the respective pathways in complement-mediated diseases. Two sandwich ELISAs detecting C1s/C1-INH and MASP-1/C1-INH complex were developed and tested thoroughly, and it was investigated whether C1s/C1-INH and MASP-1/C1-INH complexes could serve as markers for either early CP or LP activation. In addition, a reference range for these complexes in healthy adults was defined, and the assays were clinically validated utilizing samples of 414 COVID-19 patients and 96 healthy controls. The immunoassays can reliably measure C1s/C1-INH and MASP-1/C1-INH complex concentrations in EDTA plasma from healthy and diseased individuals. Both complex levels are increased in serum when activated with zymosan, making them suitable markers for early classical and early lectin pathway activation. Furthermore, measurements of C1-INH complexes in 96 healthy adults showed normally distributed C1s/C1-INH complex levels with a physiological concentration of 1846 ± 1060 ng/mL (mean ± 2SD) and right-skewed distribution of MASP-1/C1-INH complex levels with a median concentration of 36.9 (13.18 - 87.89) ng/mL (2.5-97.5 percentile range), while levels of both complexes were increased in COVID-19 patients (p<0.0001). The newly developed assays measure C1-INH complex levels in an accurate way. C1s/C1-INH and MASP-1/C1-INH complexes are suitable markers to assess early classical and lectin pathway activation. An initial reference range was set and first studies showed that these markers have added value for investigating and unraveling complement activation in human disease.
Collapse
Affiliation(s)
- Lisa Hurler
- Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
| | - Erik J. M. Toonen
- Research and Development Department, Hycult Biotech, Uden, Netherlands
| | - Erika Kajdácsi
- Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
| | - Bregje van Bree
- Research and Development Department, Hycult Biotech, Uden, Netherlands
| | | | - Wieke de Bruin
- Research and Development Department, Hycult Biotech, Uden, Netherlands
| | - Paul A. Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Laura Bergamaschi
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | | | - György Sinkovits
- Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
| | - László Cervenak
- Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
| | - Reinhard Würzner
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Zoltán Prohászka
- Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
- Research Group for Immunology and Haematology, Semmelweis University – Eötvös Loránd Research Network (Office for Supported Research Groups), Budapest, Hungary
| |
Collapse
|
18
|
Abolhassani H, Delavari S, Landegren N, Shokri S, Bastard P, Du L, Zuo F, Hajebi R, Abolnezhadian F, Iranparast S, Modaresi M, Vosughimotlagh A, Salami F, Aranda-Guillén M, Cobat A, Marcotte H, Zhang SY, Zhang Q, Rezaei N, Casanova JL, Kämpe O, Hammarström L, Pan-Hammarström Q. Genetic and immunologic evaluation of children with inborn errors of immunity and severe or critical COVID-19. J Allergy Clin Immunol 2022; 150:1059-1073. [PMID: 36113674 PMCID: PMC9472457 DOI: 10.1016/j.jaci.2022.09.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Most severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected individuals are asymptomatic or only exhibit mild disease. In about 10% of cases, the infection leads to hypoxemic pneumonia, although it is much more rare in children. OBJECTIVE We evaluated 31 young patients aged 0.5 to 19 years who had preexisting inborn errors of immunity (IEI) but lacked a molecular diagnosis and were later diagnosed with coronavirus disease 2019 (COVID-19) complications. METHODS Genetic evaluation by whole-exome sequencing was performed in all patients. SARS-CoV-2-specific antibodies, autoantibodies against type I IFN (IFN-I), and inflammatory factors in plasma were measured. We also reviewed COVID-19 disease severity/outcome in reported IEI patients. RESULTS A potential genetic cause of the IEI was identified in 28 patients (90.3%), including mutations that may affect IFN signaling, T- and B-cell function, the inflammasome, and the complement system. From tested patients 65.5% had detectable virus-specific antibodies, and 6.8% had autoantibodies neutralizing IFN-I. Five patients (16.1%) fulfilled the diagnostic criteria of multisystem inflammatory syndrome in children. Eleven patients (35.4%) died of COVID-19 complications. All together, at least 381 IEI children with COVID-19 have been reported in the literature to date. Although many patients with asymptomatic or mild disease may not have been reported, severe presentation of COVID-19 was observed in 23.6% of the published cases, and the mortality rate was 8.7%. CONCLUSIONS Young patients with preexisting IEI may have higher mortality than children without IEI when infected with SARS-CoV-2. Elucidating the genetic basis of IEI patients with severe/critical COVID-19 may help to develop better strategies for prevention and treatment of severe COVID-19 disease and complications in pediatric patients.
Collapse
Affiliation(s)
- Hassan Abolhassani
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Huddinge, Sweden; Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Samaneh Delavari
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nils Landegren
- Centre for Molecular Medicine, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden; Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Sima Shokri
- Department of Pediatrics, School of Medicine, Hazrat-e Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Paul Bastard
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris, France; University of Paris, Imagine Institute, Paris, France
| | - Likun Du
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Huddinge, Sweden
| | - Fanglei Zuo
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Huddinge, Sweden
| | - Reza Hajebi
- Department of General Surgery, School of Medicine, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Abolnezhadian
- Department of Pediatrics, Abuzar Children's Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sara Iranparast
- Department of Immunology, Faculty of Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammadreza Modaresi
- Division of Pediatrics Pulmonary Disease, Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Vosughimotlagh
- Department of Pediatrics, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Fereshte Salami
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maribel Aranda-Guillén
- Centre for Molecular Medicine, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Aurélie Cobat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris, France; University of Paris, Imagine Institute, Paris, France
| | - Harold Marcotte
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Huddinge, Sweden
| | - Shen-Ying Zhang
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris, France; University of Paris, Imagine Institute, Paris, France
| | - Qian Zhang
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris, France
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Jean-Laurent Casanova
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris, France; University of Paris, Imagine Institute, Paris, France; Howard Hughes Medical Institute, New York, NY
| | - Olle Kämpe
- Centre for Molecular Medicine, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden; Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital, Stockholm, Sweden
| | - Lennart Hammarström
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Huddinge, Sweden.
| | - Qiang Pan-Hammarström
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Huddinge, Sweden.
| |
Collapse
|
19
|
Lo MW, Amarilla AA, Lee JD, Albornoz EA, Modhiran N, Clark RJ, Ferro V, Chhabra M, Khromykh AA, Watterson D, Woodruff TM. SARS-CoV-2 triggers complement activation through interactions with heparan sulfate. Clin Transl Immunology 2022; 11:e1413. [PMID: 35999893 PMCID: PMC9387400 DOI: 10.1002/cti2.1413] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/26/2022] [Accepted: 08/08/2022] [Indexed: 12/30/2022] Open
Abstract
Objectives To determine whether SARS‐CoV‐2 can trigger complement activation, the pathways that are involved and the functional significance of the resultant effect. Methods SARS‐CoV‐2 was inoculated into a human lepirudin‐anticoagulated whole blood model, which contains a full repertoire of complement factors and leukocytes that express complement receptors. Complement activation was determined by measuring C5a production with an ELISA, and pretreatment with specific inhibitors was used to identify the pathways involved. The functional significance of this was then assessed by measuring markers of C5a signalling including leukocyte C5aR1 internalisation and CD11b upregulation with flow cytometry. Results SARS‐CoV‐2 inoculation in this whole blood model caused progressive C5a production over 24 h, which was significantly reduced by inhibitors for factor B, C3, C5 and heparan sulfate. However, this phenomenon could not be replicated in cell‐free plasma, highlighting the requirement for cell surface interactions with heparan sulfate. Functional analysis of this phenomenon revealed that C5aR1 signalling and CD11b upregulation in granulocytes and monocytes was delayed and only occurred after 24 h. Conclusion SARS‐CoV‐2 is a noncanonical alternative pathway activator that progressively triggers complement activation through interactions with heparan sulfate.
Collapse
Affiliation(s)
- Martin W Lo
- School of Biomedical Sciences, Faculty of Medicine University of Queensland Brisbane QLD Australia
| | - Alberto A Amarilla
- School of Chemistry and Molecular Biosciences University of Queensland Brisbane QLD Australia
| | - John D Lee
- School of Biomedical Sciences, Faculty of Medicine University of Queensland Brisbane QLD Australia
| | - Eduardo A Albornoz
- School of Biomedical Sciences, Faculty of Medicine University of Queensland Brisbane QLD Australia
| | - Naphak Modhiran
- School of Chemistry and Molecular Biosciences University of Queensland Brisbane QLD Australia
| | - Richard J Clark
- School of Biomedical Sciences, Faculty of Medicine University of Queensland Brisbane QLD Australia
| | - Vito Ferro
- School of Chemistry and Molecular Biosciences University of Queensland Brisbane QLD Australia.,Australian Infectious Diseases Research Centre Global Virus Network Centre of Excellence Brisbane QLD Australia
| | - Mohit Chhabra
- School of Chemistry and Molecular Biosciences University of Queensland Brisbane QLD Australia
| | - Alexander A Khromykh
- School of Chemistry and Molecular Biosciences University of Queensland Brisbane QLD Australia.,Australian Infectious Diseases Research Centre Global Virus Network Centre of Excellence Brisbane QLD Australia
| | - Daniel Watterson
- School of Chemistry and Molecular Biosciences University of Queensland Brisbane QLD Australia.,Australian Infectious Diseases Research Centre Global Virus Network Centre of Excellence Brisbane QLD Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine University of Queensland Brisbane QLD Australia.,Australian Infectious Diseases Research Centre Global Virus Network Centre of Excellence Brisbane QLD Australia
| |
Collapse
|
20
|
De Leeuw E, Van Damme KFA, Declercq J, Bosteels C, Maes B, Tavernier SJ, Detalle L, Smart T, Glatt S, Debeuf N, Deckers J, Lameire S, Vandecasteele SJ, De Neve N, Demedts IK, Govaerts E, Knoop C, Vanhove K, Moutschen M, Terryn W, Depuydt P, Van Braeckel E, Haerynck F, Hendrickx TCJ, Parrein V, Lalla M, Brittain C, Lambrecht BN. Efficacy and safety of the investigational complement C5 inhibitor zilucoplan in patients hospitalized with COVID-19: an open-label randomized controlled trial. Respir Res 2022; 23:202. [PMID: 35945604 PMCID: PMC9361275 DOI: 10.1186/s12931-022-02126-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/01/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The efficacy and safety of complement inhibition in COVID-19 patients is unclear. METHODS A multicenter randomized controlled, open-label trial. Hospitalized COVID-19 patients with signs of systemic inflammation and hypoxemia (PaO2/FiO2 below 350 mmHg) were randomized (2:1 ratio) to receive standard of care with or without the C5 inhibitor zilucoplan daily for 14 days, under antibiotic prophylaxis. The primary outcome was improvement in oxygenation at day 6 and 15. RESULTS 81 patients were randomly assigned to zilucoplan (n = 55) or the control group (n = 26). 78 patients were included in the safety and primary analysis. Most were men (87%) and the median age was 63 years. The mean improvement in PaO2/FiO2 from baseline to day 6 was 56.4 mmHg in the zilucoplan group and 20.6 mmHg in the control group (mean difference + 35.8; 95% confidence interval (CI) - 9.4 to 80.9; p = 0.12), an effect also observed at day 15. Day 28 mortality was 9% in the zilucoplan and 21% in the control group (odds ratio 0.4; 95% CI 0.1 to 1.5). At long-term follow up, the distance walked in a 6-min test was 539.7 m in zilucoplan and 490.6 m in the control group (p = 0.18). Zilucoplan lowered serum C5b-9 (p < 0.001) and interleukin-8 (p = 0.03) concentration compared with control. No relevant safety differences between the zilucoplan and control group were identified. CONCLUSION Administration of zilucoplan to COVID-19 patients in this proof-of-concept randomized trial was well tolerated under antibiotic prophylaxis. While not reaching statistical significance, indicators of respiratory function (PaO2/FiO2) and clinical outcome (mortality and 6-min walk test) suggest that C5 inhibition might be beneficial, although this requires further research in larger randomized studies.
Collapse
Affiliation(s)
- Elisabeth De Leeuw
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Karel F A Van Damme
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Jozefien Declercq
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Cedric Bosteels
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Bastiaan Maes
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Simon J Tavernier
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Primary Immunodeficiency Research Lab, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | | | | | | | - Nincy Debeuf
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Julie Deckers
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Sahine Lameire
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | | | - Nikolaas De Neve
- Department of Anesthesiology and Intensive Care Medicine, OLV Hospital, Aalst, Belgium
| | - Ingel K Demedts
- Department of Respiratory Medicine, AZ Delta Roeselare-Menen, Roeselare, Belgium
| | - Elke Govaerts
- Department of Pulmonary Medicine, AZ Sint-Lucas Gent, Ghent, Belgium
| | - Christiane Knoop
- Department of Pulmonary Medicine, CHU Erasme Université Libre de Bruxelles, Brussels, Belgium
| | - Karolien Vanhove
- Department of Pneumology and Respiratory Oncology, AZ Vesalius, Tongeren, Belgium
| | - Michel Moutschen
- Department of Infectious Diseases and General Internal Medicine, CHU Sart-Tilman, Université de Liège, Liège, Belgium
| | - Wim Terryn
- Department of General Internal Medicine and Nephrology, Jan Yperman Hospital, Ieper, Belgium
| | - Pieter Depuydt
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Intensive Care Unit, Ghent University Hospital, Ghent, Belgium
| | - Eva Van Braeckel
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Filomeen Haerynck
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Primary Immunodeficiency Research Lab, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Tine C J Hendrickx
- Clinical Trial Center, Pharmacy Department, AZ Sint-Lucas Gent, Ghent, Belgium
| | | | | | | | - Bart N Lambrecht
- Laboratory of Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium.
| |
Collapse
|
21
|
Lamerton RE, Marcial-Juarez E, Faustini SE, Perez-Toledo M, Goodall M, Jossi SE, Newby ML, Chapple I, Dietrich T, Veenith T, Shields AM, Harper L, Henderson IR, Rayes J, Wraith DC, Watson SP, Crispin M, Drayson MT, Richter AG, Cunningham AF. SARS-CoV-2 Spike- and Nucleoprotein-Specific Antibodies Induced After Vaccination or Infection Promote Classical Complement Activation. Front Immunol 2022; 13:838780. [PMID: 35860286 PMCID: PMC9289266 DOI: 10.3389/fimmu.2022.838780] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 06/07/2022] [Indexed: 12/19/2022] Open
Abstract
Antibodies specific for the spike glycoprotein (S) and nucleocapsid (N) SARS-CoV-2 proteins are typically present during severe COVID-19, and induced to S after vaccination. The binding of viral antigens by antibody can initiate the classical complement pathway. Since complement could play pathological or protective roles at distinct times during SARS-CoV-2 infection we determined levels of antibody-dependent complement activation along the complement cascade. Here, we used an ELISA assay to assess complement protein binding (C1q) and the deposition of C4b, C3b, and C5b to S and N antigens in the presence of antibodies to SARS-CoV-2 from different test groups: non-infected, single and double vaccinees, non-hospitalised convalescent (NHC) COVID-19 patients and convalescent hospitalised (ITU-CONV) COVID-19 patients. C1q binding correlates strongly with antibody responses, especially IgG1 levels. However, detection of downstream complement components, C4b, C3b and C5b shows some variability associated with the subject group from whom the sera were obtained. In the ITU-CONV, detection of C3b-C5b to S was observed consistently, but this was not the case in the NHC group. This is in contrast to responses to N, where median levels of complement deposition did not differ between the NHC and ITU-CONV groups. Moreover, for S but not N, downstream complement components were only detected in sera with higher IgG1 levels. Therefore, the classical pathway is activated by antibodies to multiple SARS-CoV-2 antigens, but the downstream effects of this activation may differ depending the disease status of the subject and on the specific antigen targeted.
Collapse
Affiliation(s)
- Rachel E. Lamerton
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Edith Marcial-Juarez
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Sian E. Faustini
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Marisol Perez-Toledo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Margaret Goodall
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Siân E. Jossi
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Maddy L. Newby
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Iain Chapple
- Periodontal Research Group, School of Dentistry, Institute of Clinical Sciences, University of Birmingham, and Birmingham Community Healthcare National Health Service Trust, Birmingham, United Kingdom
| | - Thomas Dietrich
- Periodontal Research Group, School of Dentistry, Institute of Clinical Sciences, University of Birmingham, and Birmingham Community Healthcare National Health Service Trust, Birmingham, United Kingdom
| | - Tonny Veenith
- Department of Critical Care Medicine, University Hospitals Birmingham National Health Service (NHS) Trust, Birmingham, United Kingdom
| | - Adrian M. Shields
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Lorraine Harper
- Institute of Applied Health Research, University of Birmingham, Birmingham, United Kingdom
| | - Ian R. Henderson
- Institute for Molecular Bioscience, University of Queensland, St Lucia, QLD, Australia
| | - Julie Rayes
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - David C. Wraith
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Steve P. Watson
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Max Crispin
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Mark T. Drayson
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Alex G. Richter
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Adam F. Cunningham
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
22
|
Intertwined pathways of complement activation command the pathogenesis of lupus nephritis. Transl Res 2022; 245:18-29. [PMID: 35296451 PMCID: PMC9167748 DOI: 10.1016/j.trsl.2022.03.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 12/26/2022]
Abstract
The complement system is involved in the origin of autoimmunity and systemic lupus erythematosus. Both genetic deficiency of complement components and excessive activation are involved in primary and secondary renal diseases, including lupus nephritis. Among the pathways, the classical pathway has long been accepted as the main pathway of complement activation in systemic lupus erythematosus. However, more recent studies have shown the contribution of factors B and D which implies the involvement of the alternative pathway. While there is evidence on the role of the lectin pathway in systemic lupus erythematosus, it is yet to be demonstrated whether this pathway is protective or harmful in lupus nephritis. Complement is being explored for the development of disease biomarkers and therapeutic targeting. In the current review we discuss the involvement of complement in lupus nephritis.
Collapse
|
23
|
Gavriilaki E, Tsiftsoglou SA, Touloumenidou T, Farmaki E, Panagopoulou P, Michailidou E, Koravou EE, Mavrikou I, Iosifidis E, Tsiatsiou O, Papadimitriou E, Papadopoulou-Alataki E, Papayanni PG, Varelas C, Kokkoris S, Papalexandri A, Fotoulaki M, Galli-Tsinopoulou A, Zafeiriou D, Roilides E, Sakellari I, Anagnostopoulos A, Tragiannidis A. Targeted Genotyping of MIS-C Patients Reveals a Potential Alternative Pathway Mediated Complement Dysregulation during COVID-19 Infection. Curr Issues Mol Biol 2022; 44:2811-2824. [PMID: 35877417 PMCID: PMC9325260 DOI: 10.3390/cimb44070193] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/21/2022] [Accepted: 06/24/2022] [Indexed: 11/24/2022] Open
Abstract
Complement dysregulation has been documented in adults with COVID-19 and implicated in relevant pediatric inflammatory responses against SARS-CoV-2. We propose that signatures of complement missense coding SNPs associated with dysregulation could also be identified in children with multisystem inflammatory syndrome (MIS-C). We investigated 71 pediatric patients with RT-PCR validated SARS-CoV-2 hospitalized in pediatric COVID-19 care units (November 2020-March 2021) in three major groups. Seven (7) patients suffered from MIS-C (MIS-C group), 32 suffered from COVID-19 and were hospitalized (admitted group), whereas 32 suffered from COVID-19, but were sent home. All patients survived and were genotyped for variations in the C3, C5, CFB, CFD, CFH, CFHR1, CFI, CD46, CD55, MASP1, MASP2, MBL2, COLEC11, FCN1, and FCN3 genes. Upon evaluation of the missense coding SNP distribution patterns along the three study groups, we noticed similarities, but also considerably increased frequencies of the alternative pathway (AP) associated with SNPs rs12614 CFB, rs1061170, and rs1065489 CFH in the MIS-C patients. Our analysis suggests that the corresponding substitutions potentially reduce the C3b-inactivation efficiency and promote slower and weaker AP C3bBb pre-convertase assembly on virions. Under these circumstances, the complement AP opsonization capacity may be impaired, leading to compromised immune clearance and systemic inflammation in the MIS-C syndrome.
Collapse
Affiliation(s)
- Eleni Gavriilaki
- Hematology Department & BMT Unit, G Papanicolaou Hospital, 57010 Thessaloniki, Greece; (T.T.); (E.-E.K.); (I.M.); (P.G.P.); (C.V.); (A.P.); (I.S.); (A.A.)
| | - Stefanos A. Tsiftsoglou
- Laboratory of Pharmacology, Department of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Tasoula Touloumenidou
- Hematology Department & BMT Unit, G Papanicolaou Hospital, 57010 Thessaloniki, Greece; (T.T.); (E.-E.K.); (I.M.); (P.G.P.); (C.V.); (A.P.); (I.S.); (A.A.)
| | - Evangelia Farmaki
- 1st Pediatric Department, Aristotle University of Thessaloniki, Hipporkation Hospital, 54642 Thessaloniki, Greece; (E.F.); (E.P.); (D.Z.)
| | - Paraskevi Panagopoulou
- 4th Pediatric Department, Aristotle University of Thessaloniki, Papageorgiou Hospital, 56429 Thessaloniki, Greece; (P.P.); (E.P.-A.); (M.F.)
| | - Elissavet Michailidou
- 3rd Pediatric Department, Aristotle University of Thessaloniki, Hippokration Hospital, 54642 Thessaloniki, Greece; (E.M.); (E.I.); (O.T.); (E.R.)
| | - Evaggelia-Evdoxia Koravou
- Hematology Department & BMT Unit, G Papanicolaou Hospital, 57010 Thessaloniki, Greece; (T.T.); (E.-E.K.); (I.M.); (P.G.P.); (C.V.); (A.P.); (I.S.); (A.A.)
| | - Ioulia Mavrikou
- Hematology Department & BMT Unit, G Papanicolaou Hospital, 57010 Thessaloniki, Greece; (T.T.); (E.-E.K.); (I.M.); (P.G.P.); (C.V.); (A.P.); (I.S.); (A.A.)
| | - Elias Iosifidis
- 3rd Pediatric Department, Aristotle University of Thessaloniki, Hippokration Hospital, 54642 Thessaloniki, Greece; (E.M.); (E.I.); (O.T.); (E.R.)
| | - Olga Tsiatsiou
- 3rd Pediatric Department, Aristotle University of Thessaloniki, Hippokration Hospital, 54642 Thessaloniki, Greece; (E.M.); (E.I.); (O.T.); (E.R.)
| | - Eleni Papadimitriou
- 1st Pediatric Department, Aristotle University of Thessaloniki, Hipporkation Hospital, 54642 Thessaloniki, Greece; (E.F.); (E.P.); (D.Z.)
| | - Efimia Papadopoulou-Alataki
- 4th Pediatric Department, Aristotle University of Thessaloniki, Papageorgiou Hospital, 56429 Thessaloniki, Greece; (P.P.); (E.P.-A.); (M.F.)
| | - Penelope Georgia Papayanni
- Hematology Department & BMT Unit, G Papanicolaou Hospital, 57010 Thessaloniki, Greece; (T.T.); (E.-E.K.); (I.M.); (P.G.P.); (C.V.); (A.P.); (I.S.); (A.A.)
| | - Christos Varelas
- Hematology Department & BMT Unit, G Papanicolaou Hospital, 57010 Thessaloniki, Greece; (T.T.); (E.-E.K.); (I.M.); (P.G.P.); (C.V.); (A.P.); (I.S.); (A.A.)
| | - Styliani Kokkoris
- Laboratory of Hematology and Hospital—Blood Transfusion Unit, Medical School, University General Hospital “Attikon”, NKUA, 12462 Athens, Greece;
| | - Apostolia Papalexandri
- Hematology Department & BMT Unit, G Papanicolaou Hospital, 57010 Thessaloniki, Greece; (T.T.); (E.-E.K.); (I.M.); (P.G.P.); (C.V.); (A.P.); (I.S.); (A.A.)
| | - Maria Fotoulaki
- 4th Pediatric Department, Aristotle University of Thessaloniki, Papageorgiou Hospital, 56429 Thessaloniki, Greece; (P.P.); (E.P.-A.); (M.F.)
| | - Assimina Galli-Tsinopoulou
- 2nd Pediatric Department, Aristotle University of Thessaloniki, AHEPA Hospital, 54621 Thessaloniki, Greece; (A.G.-T.); (A.T.)
| | - Dimitrios Zafeiriou
- 1st Pediatric Department, Aristotle University of Thessaloniki, Hipporkation Hospital, 54642 Thessaloniki, Greece; (E.F.); (E.P.); (D.Z.)
| | - Emmanuel Roilides
- 3rd Pediatric Department, Aristotle University of Thessaloniki, Hippokration Hospital, 54642 Thessaloniki, Greece; (E.M.); (E.I.); (O.T.); (E.R.)
| | - Ioanna Sakellari
- Hematology Department & BMT Unit, G Papanicolaou Hospital, 57010 Thessaloniki, Greece; (T.T.); (E.-E.K.); (I.M.); (P.G.P.); (C.V.); (A.P.); (I.S.); (A.A.)
| | - Achilles Anagnostopoulos
- Hematology Department & BMT Unit, G Papanicolaou Hospital, 57010 Thessaloniki, Greece; (T.T.); (E.-E.K.); (I.M.); (P.G.P.); (C.V.); (A.P.); (I.S.); (A.A.)
| | - Athanasios Tragiannidis
- 2nd Pediatric Department, Aristotle University of Thessaloniki, AHEPA Hospital, 54621 Thessaloniki, Greece; (A.G.-T.); (A.T.)
| |
Collapse
|
24
|
Govender S, Naicker T. The Contribution of Complement Protein C1q in COVID-19 and HIV Infection Comorbid with Preeclampsia: A Review. Int Arch Allergy Immunol 2022; 183:1114-1126. [PMID: 35661665 PMCID: PMC9393774 DOI: 10.1159/000524976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/21/2022] [Indexed: 11/19/2022] Open
Abstract
Dysregulation in component 1q (C1q) levels is associated with weak placental development in preeclampsia (PE). Human immunodeficiency virus infection (HIV-1) triggers the C1q complex, resulting in opsonization of healthy host cells, contributing to their removal, and augmented progression of HIV disease. In coronavirus disease 2019 (COVID-19)-infected patients, the deposition of C1q activates the complement. Considering the paucity of data, this review highlights a significant gap in the potential of C1q in the immunocompromised state of preeclamptic HIV-infected women and COVID-19 infection. In PE, C1q is downregulated; while in antiretroviral treatment-treated HIV/COVID-19 infected patients, C1q is upregulated. It is plausible that C1q is augmented in the triad and may exacerbate severity of disease. This thereby provides a foundation for future intended research which involves the investigation of single nucleotide polymorphism expression of the C1q gene, specifically in these diseases.
Collapse
Affiliation(s)
- Sumeshree Govender
- Optics & Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Thajasvarie Naicker
- Optics & Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
25
|
Jiménez D, Torres Arias M. Immunouniverse of SARS-CoV-2. Immunol Med 2022; 45:186-224. [PMID: 35502127 DOI: 10.1080/25785826.2022.2066251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
SARS-CoV-2 virus has become a global health problem that has caused millions of deaths worldwide. The infection can present with multiple clinical features ranging from asymptomatic or mildly symptomatic patients to patients with severe or critical illness that can even lead to death. Although the immune system plays an important role in pathogen control, SARS-CoV-2 can drive dysregulation of this response and trigger severe immunopathology. Exploring the mechanisms of the immune response involved in host defense against SARS-CoV-2 allows us to understand its immunopathogenesis and possibly detect features that can be used as potential therapies to eliminate the virus. The main objective of this review on SARS-CoV-2 is to highlight the interaction between the virus and the immune response. We explore the function and action of the immune system, the expression of molecules at the site of infection that cause hyperinflammation and hypercoagulation disorders, the factors leading to the development of pneumonia and subsequent severe acute respiratory distress syndrome which is the leading cause of death in patients with COVID-19.
Collapse
Affiliation(s)
- Dennis Jiménez
- Departamento de Ciencias de la Vida y Agricultura, Carrera de Ingeniería en Biotecnología, Universidad de las Fuerzas Armadas ESPE, Sangolquí, Pichincha, Ecuador
| | - Marbel Torres Arias
- Departamento de Ciencias de la Vida y Agricultura, Carrera de Ingeniería en Biotecnología, Universidad de las Fuerzas Armadas ESPE, Sangolquí, Pichincha, Ecuador.,Laboratorio de Inmunología y Virología, CENCINAT, GISAH, Universidad de las Fuerzas Armadas, Sangolquí, Pichincha, Ecuador
| |
Collapse
|
26
|
Non-invasive diagnostic potential of microRNA-203 in liquid biopsy of urothelial carcinoma of bladder. Mol Cell Biochem 2022; 477:2173-2182. [PMID: 35445913 PMCID: PMC9021364 DOI: 10.1007/s11010-022-04431-2] [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: 01/25/2022] [Accepted: 04/01/2022] [Indexed: 11/07/2022]
Abstract
Increased CD44 antigen activity has been reported in recurrent cases of UBC. To date, no reliable biomarker is available with high significance and specificity for non-invasive detection of UBC. This study aimed to identify a CD44-linked microRNAs (miRNAs) (miR-9, miR-34a, miR-203) for non-invasive diagnosis of bladder cancer from other urinary tract malignancies. The expression of CD44-linked miRNAs was examined in serum, urine, and tissue specimens of Indian UBC patients (N = 25). For this purpose, healthy subjects (N = 25) and benign prostatic hyperplasia (BPH) (N = 10) patients were taken as controls. The relative expression of miRNAs was analyzed in serum, urine, and tissue samples using real-time quantitative reverse transcription PCR (qRT-PCR). The diagnostic potential of these miRNAs was accessed by plotting ROC curve. Increased miR-9 expression was observed in serum of UBC patients than healthy and BPH controls. In UBC patients, miR-34a expression was lower than healthy controls but non-significant as compared to BPH. miR-203 expression was considerably higher in serum of UBC patients but non-significant as compared to BPH controls. miR-203 was found to be considerably higher in urine samples from UBC patients as compared to BPH and healthy controls. The diagnostic potential of these miRNAs was evaluated using the ROC curve. Higher miR-203 levels in the urine of Indian UBC patients demonstrate its non-invasive diagnostic ability out of the three miRNAs studied. Our results characterize the non-invasive diagnostic potential of CD44-linked miR-203 in the urine of Indian UBC patients, which could be utilized in clinical settings in future after validation in larger patient cohort.
Collapse
|
27
|
Georg P, Astaburuaga-García R, Bonaguro L, Brumhard S, Michalick L, Lippert LJ, Kostevc T, Gäbel C, Schneider M, Streitz M, Demichev V, Gemünd I, Barone M, Tober-Lau P, Helbig ET, Hillus D, Petrov L, Stein J, Dey HP, Paclik D, Iwert C, Mülleder M, Aulakh SK, Djudjaj S, Bülow RD, Mei HE, Schulz AR, Thiel A, Hippenstiel S, Saliba AE, Eils R, Lehmann I, Mall MA, Stricker S, Röhmel J, Corman VM, Beule D, Wyler E, Landthaler M, Obermayer B, von Stillfried S, Boor P, Demir M, Wesselmann H, Suttorp N, Uhrig A, Müller-Redetzky H, Nattermann J, Kuebler WM, Meisel C, Ralser M, Schultze JL, Aschenbrenner AC, Thibeault C, Kurth F, Sander LE, Blüthgen N, Sawitzki B. Complement activation induces excessive T cell cytotoxicity in severe COVID-19. Cell 2022; 185:493-512.e25. [PMID: 35032429 PMCID: PMC8712270 DOI: 10.1016/j.cell.2021.12.040] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/22/2021] [Accepted: 12/22/2021] [Indexed: 12/20/2022]
Abstract
Severe COVID-19 is linked to both dysfunctional immune response and unrestrained immunopathology, and it remains unclear whether T cells contribute to disease pathology. Here, we combined single-cell transcriptomics and single-cell proteomics with mechanistic studies to assess pathogenic T cell functions and inducing signals. We identified highly activated CD16+ T cells with increased cytotoxic functions in severe COVID-19. CD16 expression enabled immune-complex-mediated, T cell receptor-independent degranulation and cytotoxicity not found in other diseases. CD16+ T cells from COVID-19 patients promoted microvascular endothelial cell injury and release of neutrophil and monocyte chemoattractants. CD16+ T cell clones persisted beyond acute disease maintaining their cytotoxic phenotype. Increased generation of C3a in severe COVID-19 induced activated CD16+ cytotoxic T cells. Proportions of activated CD16+ T cells and plasma levels of complement proteins upstream of C3a were associated with fatal outcome of COVID-19, supporting a pathological role of exacerbated cytotoxicity and complement activation in COVID-19.
Collapse
Affiliation(s)
- Philipp Georg
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Rosario Astaburuaga-García
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany; IRI Life Sciences, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lorenzo Bonaguro
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany; Systems Medicine, Deutsches Zentrum für Neurodegenerativen Erkrankungen (DZNE), Bonn, Germany
| | - Sophia Brumhard
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Laura Michalick
- Institute of Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Lena J Lippert
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Tomislav Kostevc
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christiane Gäbel
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Maria Schneider
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Mathias Streitz
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Vadim Demichev
- Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, UK; Department of Biochemistry, Charité - Universitätsmedizin Berlin, Berlin, Germany; Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge, UK
| | - Ioanna Gemünd
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany; PRECISE Platform for Genomics and Epigenomics at DZNE, University of Bonn, Bonn, Germany; Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Matthias Barone
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Pinkus Tober-Lau
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Elisa T Helbig
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - David Hillus
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lev Petrov
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Julia Stein
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Hannah-Philine Dey
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Daniela Paclik
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christina Iwert
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Mülleder
- Core Facility, High Throughput Mass Spectrometry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Simran Kaur Aulakh
- Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, UK
| | - Sonja Djudjaj
- Institute of Pathology, University Clinic Aachen, RWTH Aachen, Aachen, Germany
| | - Roman D Bülow
- Institute of Pathology, University Clinic Aachen, RWTH Aachen, Aachen, Germany
| | - Henrik E Mei
- Mass Cytometry Laboratory, DRFZ Berlin, A Leibniz Institute, Berlin, Germany
| | - Axel R Schulz
- Mass Cytometry Laboratory, DRFZ Berlin, A Leibniz Institute, Berlin, Germany
| | - Andreas Thiel
- Si-M/"Der Simulierte Mensch" a Science Framework of Technische Universität Berlin and Charité - Universitätsmedizin Berlin, Berlin, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt - Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Stefan Hippenstiel
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Center for Infection Research (HZI), Würzburg, Germany
| | - Roland Eils
- Center for Digital Health, Berlin Institute of Health (BIH), Charité - Universitätsmedizin Berlin, Berlin, Germany; German Center for Lung Research (DZL), Berlin, Germany
| | - Irina Lehmann
- Center for Digital Health, Berlin Institute of Health (BIH), Charité - Universitätsmedizin Berlin, Berlin, Germany; German Center for Lung Research (DZL), Berlin, Germany
| | - Marcus A Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany; German Center for Lung Research (DZL), Associated Partner, Berlin, Germany; Berlin Institute of Health (BIH), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Stricker
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jobst Röhmel
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Victor M Corman
- Institute of Virology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health (BIH), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Emanuel Wyler
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin Institute for Medical Systems Biology, Berlin, Germany
| | - Markus Landthaler
- IRI Life Sciences, Humboldt-Universität zu Berlin, Berlin, Germany; Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin Institute for Medical Systems Biology, Berlin, Germany
| | - Benedikt Obermayer
- Core Unit Bioinformatics, Berlin Institute of Health (BIH), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Peter Boor
- Institute of Pathology, University Clinic Aachen, RWTH Aachen, Aachen, Germany; Department of Nephrology, University Clinic Aachen, RWTH Aachen, Aachen, Germany; Electron Microscopy Facility, University Clinic Aachen, RWTH Aachen, Aachen, Germany
| | - Münevver Demir
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Hans Wesselmann
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Norbert Suttorp
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany; German Center for Lung Research (DZL), Gießen, Germany
| | - Alexander Uhrig
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Holger Müller-Redetzky
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jacob Nattermann
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Meisel
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany; Department of Immunology, Labor Berlin, Charité Vivantes, Berlin, Germany
| | - Markus Ralser
- Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, UK; Department of Biochemistry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Joachim L Schultze
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany; Systems Medicine, Deutsches Zentrum für Neurodegenerativen Erkrankungen (DZNE), Bonn, Germany; PRECISE Platform for Genomics and Epigenomics at DZNE, University of Bonn, Bonn, Germany
| | - Anna C Aschenbrenner
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany; Systems Medicine, Deutsches Zentrum für Neurodegenerativen Erkrankungen (DZNE), Bonn, Germany; PRECISE Platform for Genomics and Epigenomics at DZNE, University of Bonn, Bonn, Germany; Department of Internal Medicine, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Charlotte Thibeault
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Florian Kurth
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany; Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine, Department of Medicine I, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Leif E Sander
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nils Blüthgen
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany; IRI Life Sciences, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Birgit Sawitzki
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany; Berlin Institute of Health (BIH), Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
28
|
Niederreiter J, Eck C, Ries T, Hartmann A, Märkl B, Büttner-Herold M, Amann K, Daniel C. Complement Activation via the Lectin and Alternative Pathway in Patients With Severe COVID-19. Front Immunol 2022; 13:835156. [PMID: 35237273 PMCID: PMC8884149 DOI: 10.3389/fimmu.2022.835156] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/11/2022] [Indexed: 01/08/2023] Open
Abstract
Complement plays an important role in the direct defense to pathogens, but can also activate immune cells and the release of pro-inflammatory cytokines. However, in critically ill patients with COVID-19 the immune system is inadequately activated leading to severe acute respiratory syndrome (SARS) and acute kidney injury, which is associated with higher mortality. Therefore, we characterized local complement deposition as a sign of activation in both lungs and kidneys from patients with severe COVID-19. Using immunohistochemistry we investigated deposition of complement factors C1q, MASP-2, factor D (CFD), C3c, C3d and C5b-9 as well as myeloperoxidase (MPO) positive neutrophils and SARS-CoV-2 virus particles in lungs and kidneys from 38 patients who died from COVID-19. In addition, tissue damage was analyzed using semi-quantitative scores followed by correlation with complement deposition. Autopsy material from non-COVID patients who died from cardiovascular causes, cerebral hemorrhage and pulmonary embolism served as control (n=8). Lung injury in samples from COVID-19 patients was significantly more pronounced compared to controls with formation of hyaline membranes, thrombi and edema. In addition, in the kidney tubular injury was higher in these patients and correlated with lung injury (r=0.361*). In autopsy samples SARS-CoV-2 spike protein was detected in 22% of the lungs of COVID-19 patients but was lacking in kidneys. Complement activation was significantly stronger in lung samples from patients with COVID-19 via the lectin and alternative pathway as indicated by deposition of MASP-2, CFD, C3d and C5b9. Deposits in the lung were predominantly detected along the alveolar septa, the hyaline membranes and in the alveolar lumina. In the kidney, complement was significantly more deposited in patients with COVID-19 in peritubular capillaries and tubular basement membranes. Renal COVID-19-induced complement activation occurred via the lectin pathway, while activation of the alternative pathway was similar in both groups. Furthermore, MPO-positive neutrophils were found in significantly higher numbers in lungs and kidneys of COVID-19 patients and correlated with local MASP-2 deposition. In conclusion, in patients who died from SARS-CoV-2 infection complement was activated in both lungs and kidneys indicating that complement might be involved in systemic worsening of the inflammatory response. Complement inhibition might thus be a promising treatment option to prevent deregulated activation and subsequent collateral tissue injury in COVID-19.
Collapse
Affiliation(s)
- Janina Niederreiter
- Department of Nephropathology, University Hospital Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Christine Eck
- Department of Nephropathology, University Hospital Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Tajana Ries
- Department of Nephropathology, University Hospital Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Bruno Märkl
- General Pathology and Molecular Diagnostics, Medical Faculty Augsburg, University Augsburg, Augsburg, Germany
| | - Maike Büttner-Herold
- Department of Nephropathology, University Hospital Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Kerstin Amann
- Department of Nephropathology, University Hospital Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Christoph Daniel
- Department of Nephropathology, University Hospital Erlangen, Friedrich-Alexander-University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
29
|
Abstract
Hyperactivation of the complement and coagulation systems is recognized as part of the clinical syndrome of COVID-19. Here we review systemic complement activation and local complement activation in response to the causative virus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and their currently known relationships to hyperinflammation and thrombosis. We also provide an update on early clinical findings and emerging clinical trial evidence that suggest potential therapeutic benefit of complement inhibition in severe COVID-19.
Collapse
Affiliation(s)
- Behdad Afzali
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Marina Noris
- Istituto di Ricerche Farmacologiche "Mario Negri", Clinical Research Center for Rare Diseases "Aldo e Cele Daccò", Ranica, Italy.
- "Centro Anna Maria Astori", Bergamo, Italy.
| | - Bart N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium.
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, Netherlands.
| | - Claudia Kemper
- Complement and Inflammation Research Section (CIRS), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany.
| |
Collapse
|
30
|
Singh A, Gupta N, Khandakar H, Kaushal S, Seth A, Pandey RM, Sharma A. Autophagy-associated HMGB-1 as a novel potential circulating non-invasive diagnostic marker for detection of Urothelial Carcinoma of Bladder. Mol Cell Biochem 2022; 477:493-505. [PMID: 34796446 PMCID: PMC8601373 DOI: 10.1007/s11010-021-04299-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/04/2021] [Indexed: 01/04/2023]
Abstract
Urothelial carcinoma of bladder (UBC), a highly prevalent urological malignancy associated with high mortality and recurrence rate. Standard diagnostic method currently being used is cystoscopy but its invasive nature and low sensitivity stresses for identifying predictive diagnostic marker. Autophagy, a cellular homeostasis maintaining process, is usually dysregulated in cancer and its role is still enigmatic in UBC. In this study, 30 UBC patients and healthy controls were enrolled. Histopathologically confirmed tumor and adjacent normal tissue were acquired from patients. Molecular expression and tissue localization of autophagy-associated molecules (HMGB-1, RAGE, beclin, LC-3, and p62) were investigated. Serum HMGB-1 concentration was measured in UBC patients and healthy controls. ROC curves were plotted to evaluate diagnostic potential. Transcript, protein, and IHC expression of HMGB-1, RAGE, beclin, and LC-3 displayed upregulated expression, while p62 was downregulated in bladder tumor tissue. Serum HMGB-1 levels were elevated in UBC patients. Transcript and circulatory levels of HMGB-1 showed positive correlation and displayed a positive trend with disease severity. Upon comparison with clinicopathological parameters, HMGB-1 emerged as molecule of statistical significance to exhibit association. HMGB-1 exhibited optimum sensitivity and specificity in serum. The positive correlation between tissue and serum levels of HMGB-1 showcases serum as a representation of in situ scenario, suggesting its clinical applicability for non-invasive testing. Moreover, optimum sensitivity and specificity displayed by HMGB-1 along with significant association with clinicopathological parameters makes it a potential candidate to be used as diagnostic marker for early detection of UBC but requires further validation in larger cohort.
Collapse
Affiliation(s)
- Aishwarya Singh
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Nidhi Gupta
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Hena Khandakar
- Department of Pathology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Seema Kaushal
- Department of Pathology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Amlesh Seth
- Department of Urology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - R M Pandey
- Department of Biostatistics, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Alpana Sharma
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi, India.
| |
Collapse
|
31
|
Asteris PG, Gavriilaki E, Touloumenidou T, Koravou E, Koutra M, Papayanni PG, Pouleres A, Karali V, Lemonis ME, Mamou A, Skentou AD, Papalexandri A, Varelas C, Chatzopoulou F, Chatzidimitriou M, Chatzidimitriou D, Veleni A, Rapti E, Kioumis I, Kaimakamis E, Bitzani M, Boumpas D, Tsantes A, Sotiropoulos D, Papadopoulou A, Kalantzis IG, Vallianatou LA, Armaghani DJ, Cavaleri L, Gandomi AH, Hajihassani M, Hasanipanah M, Koopialipoor M, Lourenço PB, Samui P, Zhou J, Sakellari I, Valsami S, Politou M, Kokoris S, Anagnostopoulos A. Genetic prediction of ICU hospitalization and mortality in COVID‐19 patients using artificial neural networks. J Cell Mol Med 2022; 26:1445-1455. [PMID: 35064759 PMCID: PMC8899198 DOI: 10.1111/jcmm.17098] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/18/2021] [Accepted: 11/23/2021] [Indexed: 12/13/2022] Open
Abstract
There is an unmet need of models for early prediction of morbidity and mortality of Coronavirus disease‐19 (COVID‐19). We aimed to a) identify complement‐related genetic variants associated with the clinical outcomes of ICU hospitalization and death, b) develop an artificial neural network (ANN) predicting these outcomes and c) validate whether complement‐related variants are associated with an impaired complement phenotype. We prospectively recruited consecutive adult patients of Caucasian origin, hospitalized due to COVID‐19. Through targeted next‐generation sequencing, we identified variants in complement factor H/CFH, CFB, CFH‐related, CFD, CD55, C3, C5, CFI, CD46, thrombomodulin/THBD, and A Disintegrin and Metalloproteinase with Thrombospondin motifs (ADAMTS13). Among 381 variants in 133 patients, we identified 5 critical variants associated with severe COVID‐19: rs2547438 (C3), rs2250656 (C3), rs1042580 (THBD), rs800292 (CFH) and rs414628 (CFHR1). Using age, gender and presence or absence of each variant, we developed an ANN predicting morbidity and mortality in 89.47% of the examined population. Furthermore, THBD and C3a levels were significantly increased in severe COVID‐19 patients and those harbouring relevant variants. Thus, we reveal for the first time an ANN accurately predicting ICU hospitalization and death in COVID‐19 patients, based on genetic variants in complement genes, age and gender. Importantly, we confirm that genetic dysregulation is associated with impaired complement phenotype.
Collapse
Affiliation(s)
- Panagiotis G. Asteris
- Computational Mechanics Laboratory School of Pedagogical and Technological Education Athens Greece
| | - Eleni Gavriilaki
- Hematology Department BMT Unit G Papanicolaou Hospital Thessaloniki Greece
| | | | | | - Maria Koutra
- Hematology Department BMT Unit G Papanicolaou Hospital Thessaloniki Greece
| | | | | | - Vassiliki Karali
- Rheumatology and Clinical Immunology Unit University General Hospital ‘Attikon’ Athens Greece
| | - Minas E. Lemonis
- Computational Mechanics Laboratory School of Pedagogical and Technological Education Athens Greece
| | - Anna Mamou
- Computational Mechanics Laboratory School of Pedagogical and Technological Education Athens Greece
| | - Athanasia D. Skentou
- Computational Mechanics Laboratory School of Pedagogical and Technological Education Athens Greece
| | | | - Christos Varelas
- Hematology Department BMT Unit G Papanicolaou Hospital Thessaloniki Greece
| | | | - Maria Chatzidimitriou
- Biomedical Sciences Alexander Campus International Hellenic University Thessaloniki Greece
| | | | - Anastasia Veleni
- Infectious Disease Committee G Papanicolaou Hospital Thessaloniki Greece
| | - Evdoxia Rapti
- Laboratory of Hematology and Hospital Blood Transfusion Department University General Hospital ‘Attikon’NKUAMedical School Athens Greece
| | - Ioannis Kioumis
- Respiratory Failure Department G Papanicolaou HospitalAristotle University of Thessaloniki Thessaloniki Greece
| | | | - Milly Bitzani
- 1 Intensive Care Unit G Papanicolaou Hospital Thessaloniki Greece
| | - Dimitrios Boumpas
- Rheumatology and Clinical Immunology Unit University General Hospital ‘Attikon’ Athens Greece
| | - Argyris Tsantes
- Laboratory of Hematology and Hospital Blood Transfusion Department University General Hospital ‘Attikon’NKUAMedical School Athens Greece
| | | | | | | | - Lydia A. Vallianatou
- Cath and EP Laboratory/Covid Intensive Care Unit Hellenic Red Cross Hospital Athens Greece
| | - Danial J. Armaghani
- Department of Civil Engineering Faculty of Engineering University of Malaya Kuala Lumpur Malaysia
| | - Liborio Cavaleri
- Department of Civil, Environmental, Aerospace and Materials Engineering University of Palermo Palermo Italy
| | - Amir H. Gandomi
- Faculty of Engineering & Information Technology University of Technology Sydney Ultimo NSW Australia
| | - Mohsen Hajihassani
- Department of Mining Engineering Faculty of Engineering Urmia University Urmia Iran
| | - Mahdi Hasanipanah
- Institute of Research and Development Duy Tan University Da Nang Vietnam
| | | | - Paulo B. Lourenço
- Department of Civil Engineering ISISEUniversity of Minho Guimarães Portugal
| | - Pijush Samui
- Department of Civil Engineering National Institute of Technology Patna Patna Bihar India
| | - Jian Zhou
- School of Resources and Safety Engineering Central South University Changsha China
| | - Ioanna Sakellari
- Hematology Department BMT Unit G Papanicolaou Hospital Thessaloniki Greece
| | - Serena Valsami
- Hematology Laboratory ‐ Blood Bank Aretaieion HospitalSchool of MedicineNKUA Athens Greece
| | - Marianna Politou
- Hematology Laboratory ‐ Blood Bank Aretaieion HospitalSchool of MedicineNKUA Athens Greece
| | - Styliani Kokoris
- Laboratory of Hematology and Hospital Blood Transfusion Department University General Hospital ‘Attikon’NKUAMedical School Athens Greece
| | | |
Collapse
|
32
|
Liu Y, Budylowski P, Dong S, Li Z, Goroshko S, Leung LYT, Grunebaum E, Campisi P, Propst EJ, Wolter NE, Rini JM, Zia A, Ostrowski M, Ehrhardt GRA. SARS-CoV-2-Reactive Mucosal B Cells in the Upper Respiratory Tract of Uninfected Individuals. THE JOURNAL OF IMMUNOLOGY 2021; 207:2581-2588. [PMID: 34607939 DOI: 10.4049/jimmunol.2100606] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/02/2021] [Indexed: 12/15/2022]
Abstract
SARS-CoV-2 is a respiratory pathogen that can cause severe disease in at-risk populations but results in asymptomatic infections or a mild course of disease in the majority of cases. We report the identification of SARS-CoV-2-reactive B cells in human tonsillar tissue obtained from children who were negative for coronavirus disease 2019 prior to the pandemic and the generation of mAbs recognizing the SARS-CoV-2 Spike protein from these B cells. These Abs showed reduced binding to Spike proteins of SARS-CoV-2 variants and did not recognize Spike proteins of endemic coronaviruses, but subsets reacted with commensal microbiota and exhibited SARS-CoV-2-neutralizing potential. Our study demonstrates pre-existing SARS-CoV-2-reactive Abs in various B cell populations in the upper respiratory tract lymphoid tissue that may lead to the rapid engagement of the pathogen and contribute to prevent manifestations of symptomatic or severe disease.
Collapse
Affiliation(s)
- Yanling Liu
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | | | - Shilan Dong
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Zhijie Li
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Sofiya Goroshko
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Leslie Y T Leung
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Eyal Grunebaum
- Division of Immunology and Allergy, The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - Paolo Campisi
- Department of Otolaryngology-Head & Neck Surgery, The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - Evan J Propst
- Department of Otolaryngology-Head & Neck Surgery, The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - Nikolas E Wolter
- Department of Otolaryngology-Head & Neck Surgery, The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - James M Rini
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.,Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada; and
| | - Amin Zia
- dYcode.bio, Toronto, Ontario, Canada
| | - Mario Ostrowski
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Götz R A Ehrhardt
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada;
| |
Collapse
|
33
|
Rahman MH, Rana HK, Peng S, Kibria MG, Islam MZ, Mahmud SMH, Moni MA. Bioinformatics and system biology approaches to identify pathophysiological impact of COVID-19 to the progression and severity of neurological diseases. Comput Biol Med 2021; 138:104859. [PMID: 34601390 PMCID: PMC8483812 DOI: 10.1016/j.compbiomed.2021.104859] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/21/2021] [Accepted: 09/06/2021] [Indexed: 02/06/2023]
Abstract
The Coronavirus Disease 2019 (COVID-19) still tends to propagate and increase the occurrence of COVID-19 across the globe. The clinical and epidemiological analyses indicate the link between COVID-19 and Neurological Diseases (NDs) that drive the progression and severity of NDs. Elucidating why some patients with COVID-19 influence the progression of NDs and patients with NDs who are diagnosed with COVID-19 are becoming increasingly sick, although others are not is unclear. In this research, we investigated how COVID-19 and ND interact and the impact of COVID-19 on the severity of NDs by performing transcriptomic analyses of COVID-19 and NDs samples by developing the pipeline of bioinformatics and network-based approaches. The transcriptomic study identified the contributing genes which are then filtered with cell signaling pathway, gene ontology, protein-protein interactions, transcription factor, and microRNA analysis. Identifying hub-proteins using protein-protein interactions leads to the identification of a therapeutic strategy. Additionally, the incorporation of comorbidity interactions score enhances the identification beyond simply detecting novel biological mechanisms involved in the pathophysiology of COVID-19 and its NDs comorbidities. By computing the semantic similarity between COVID-19 and each of the ND, we have found gene-based maximum semantic score between COVID-19 and Parkinson's disease, the minimum semantic score between COVID-19 and Multiple sclerosis. Similarly, we have found gene ontology-based maximum semantic score between COVID-19 and Huntington disease, minimum semantic score between COVID-19 and Epilepsy disease. Finally, we validated our findings using gold-standard databases and literature searches to determine which genes and pathways had previously been associated with COVID-19 and NDs.
Collapse
Affiliation(s)
- Md Habibur Rahman
- Dept. of Computer Science and Engineering, Islamic University, Kushtia 7003, Bangladesh
| | - Humayan Kabir Rana
- Dept. of Computer Science and Engineering, Green University of Bangladesh, Dhaka, Bangladesh
| | - Silong Peng
- Institute of Automation, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Md Golam Kibria
- Dept. of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, Canada
| | - Md Zahidul Islam
- Department of Electronics, Graduate School of Engineering, Nagoya University, Japan
| | - S M Hasan Mahmud
- Dept. of Computer Science, American International University Bangladesh, Dhaka, Bangladesh
| | - Mohammad Ali Moni
- School of Health and Rehabilitation Sciences, Faculty of Health and Behavioural Sciences, The University of Queensland, St Lucia, QLD 4072, Australia.
| |
Collapse
|
34
|
Defendi F, Leroy C, Epaulard O, Clavarino G, Vilotitch A, Le Marechal M, Jacob MC, Raskovalova T, Pernollet M, Le Gouellec A, Bosson JL, Poignard P, Roustit M, Thielens N, Dumestre-Pérard C, Cesbron JY. Complement Alternative and Mannose-Binding Lectin Pathway Activation Is Associated With COVID-19 Mortality. Front Immunol 2021; 12:742446. [PMID: 34567008 PMCID: PMC8461024 DOI: 10.3389/fimmu.2021.742446] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/23/2021] [Indexed: 12/19/2022] Open
Abstract
Background The SARS-CoV-2 infection triggers excessive immune response resulting in increased levels of pro-inflammatory cytokines, endothelial injury, and intravascular coagulopathy. The complement system (CS) activation participates to this hyperinflammatory response. However, it is still unclear which activation pathways (classical, alternative, or lectin pathway) pilots the effector mechanisms that contribute to critical illness. To better understand the immune correlates of disease severity, we performed an analysis of CS activation pathways and components in samples collected from COVID-19 patients hospitalized in Grenoble Alpes University Hospital between 1 and 30 April 2020 and of their relationship with the clinical outcomes. Methods We conducted a retrospective, single-center study cohort in 74 hospitalized patients with RT-PCR-proven COVID-19. The functional activities of classical, alternative, and mannose-binding lectin (MBL) pathways and the antigenic levels of the individual components C1q, C4, C3, C5, Factor B, and MBL were measured in patients' samples during hospital admission. Hierarchical clustering with the Ward method was performed in order to identify clusters of patients with similar characteristics of complement markers. Age was included in the model. Then, the clusters were compared with the patient clinical features: rate of intensive care unit (ICU) admission, corticoid treatment, oxygen requirement, and mortality. Results Four clusters were identified according to complement parameters. Among them, two clusters revealed remarkable profiles: in one cluster (n = 15), patients exhibited activation of alternative and lectin pathways and low antigenic levels of MBL, C4, C3, Factor B, and C5 compared to all the other clusters; this cluster had the higher proportion of patients who died (27%) and required oxygen support (80%) or ICU care (53%). In contrast, the second cluster (n = 19) presented inflammatory profile with high classical pathway activity and antigenic levels of complement components; a low proportion of patients required ICU care (26%) and no patient died in this group. Conclusion These findings argue in favor of prominent activation of the alternative and MBL complement pathways in severe COVID-19, but the spectrum of complement involvement seems to be heterogeneous requiring larger studies.
Collapse
Affiliation(s)
- Federica Defendi
- Laboratoire d’Immunologie, Institut de Biologie et Pathologie, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Corentin Leroy
- Cellule d’Ingénierie des Données, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
- Centre d’Investigation Clinique de l’Innovation et de la Technologie (CIC-IT), Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Olivier Epaulard
- Service des Maladies Infectieuses et Tropicales, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
- Université Grenoble Alpes, TIMC-IMAG, Grenoble, France
| | - Giovanna Clavarino
- Laboratoire d’Immunologie, Institut de Biologie et Pathologie, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Antoine Vilotitch
- Cellule d’Ingénierie des Données, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Marion Le Marechal
- Service des Maladies Infectieuses et Tropicales, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Marie-Christine Jacob
- Laboratoire d’Immunologie, Institut de Biologie et Pathologie, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Tatiana Raskovalova
- Laboratoire d’Immunologie, Institut de Biologie et Pathologie, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Martine Pernollet
- Laboratoire d’Immunologie, Institut de Biologie et Pathologie, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Audrey Le Gouellec
- Université Grenoble Alpes, TIMC-IMAG, Grenoble, France
- Laboratoire de Biochimie, Institut de Biologie et Pathologie, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | | | - Pascal Poignard
- Université Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale (IBS), Grenoble, France
- Laboratoire de Virologie, Institut de Biologie et Pathologie, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Matthieu Roustit
- Département de Pharmacologie Clinique INSERM CIC 1406, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
- Université Grenoble Alpes, UMR 1042-HP2, INSERM, Grenoble, France
| | - Nicole Thielens
- Université Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale (IBS), Grenoble, France
| | - Chantal Dumestre-Pérard
- Laboratoire d’Immunologie, Institut de Biologie et Pathologie, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
- Université Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale (IBS), Grenoble, France
| | - Jean-Yves Cesbron
- Laboratoire d’Immunologie, Institut de Biologie et Pathologie, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| |
Collapse
|
35
|
Stack M, Sacco K, Castagnoli R, Livinski AA, Notarangelo LD, Lionakis MS. BTK inhibitors for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2): A systematic review. Clin Immunol 2021; 230:108816. [PMID: 34352390 PMCID: PMC8327577 DOI: 10.1016/j.clim.2021.108816] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/30/2021] [Accepted: 07/31/2021] [Indexed: 12/15/2022]
Abstract
Introduction The Bruton tyrosine kinase (BTK) regulates B cell and macrophage signaling, development, survival, and activation. Inhibiting BTK has been hypothesized to ameliorate lung injury in patients with severe COVID-19, however clinical outcome data is inconclusive. Objective To evaluate the clinical outcomes of BTK inhibitors (BTKinibs) in patients with COVID-19. Evidence review We searched PubMed, Embase, and Web of Science:Core on December 30, 2020. Clinical studies with at least 5 COVID-19 patients treated with BTKinibs were included. Case reports and reviews were excluded. Findings 125 articles were identified, 6 of which met inclusion criteria. The most common clinical outcomes measured were oxygen requirements (4/6) and hospitalization rate or duration (3/6). Three studies showed decreased oxygen requirements in patients who started or continued BTKinibs. All three studies that evaluated hospitalization rate or duration found favorable outcomes in those on BTKinibs. Conclusions and relevance BTKinib use was associated with decreased oxygen requirements and decreased hospitalization rates and duration.
Collapse
Affiliation(s)
- Michael Stack
- Laboratory of Clinical Immunology and Microbiology, Immunodeficiency Genetics Section, NIAID, NIH, Bethesda, MD, USA
| | - Keith Sacco
- Laboratory of Clinical Immunology and Microbiology, Immunodeficiency Genetics Section, NIAID, NIH, Bethesda, MD, USA.
| | - Riccardo Castagnoli
- Laboratory of Clinical Immunology and Microbiology, Immunodeficiency Genetics Section, NIAID, NIH, Bethesda, MD, USA
| | - Alicia A Livinski
- Division of Library Services, Office of Research Services, National Institutes of Health, Bethesda, MD, USA
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, Immunodeficiency Genetics Section, NIAID, NIH, Bethesda, MD, USA
| | - Michail S Lionakis
- Laboratory of Clinical Immunology and Microbiology, Fungal Pathogenesis Section, NIAID, NIH, Bethesda, MD, USA
| |
Collapse
|
36
|
Exosomal DNMT1 mRNA transcript is elevated in acute lymphoblastic leukemia which might reprograms leukemia progression. Cancer Genet 2021; 260-261:57-64. [PMID: 34426211 DOI: 10.1016/j.cancergen.2021.07.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/09/2021] [Accepted: 07/29/2021] [Indexed: 02/06/2023]
Abstract
DNMT1 (DNA-methyltransferase 1) is an enzyme which contributes to the process of normal embryonic development, and aberrant expression of DNMT1 leads to tumor/leukemia progression by inducing significant changes in DNA methylation and epigenetics. We found that DNMT1 mRNA transcript is elevated in Exo-PALL compared to Exo-HD. We also confirmed and showed heightened levels of DNMT1 mRNA transcript in Exo-CM of leukemia cell lines. Co-culture of Exo-PALL with target cells (leukemia B cells) showed transfer of exosomal DNMT1 mRNA transcript into the target cells, which may reprogram the biological nature of normal healthy cells and leukemia cells.
Collapse
|
37
|
Wang G, Deng J, Li J, Wu C, Dong H, Wu S, Zhong Y. The Role of High-Density Lipoprotein in COVID-19. Front Pharmacol 2021; 12:720283. [PMID: 34335279 PMCID: PMC8322438 DOI: 10.3389/fphar.2021.720283] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/06/2021] [Indexed: 01/08/2023] Open
Abstract
The current Coronavirus disease 2019 (COVID-19) pandemic has become a global challenge. Managing a large number of acutely ill patients in a short time, whilst reducing the fatality rate and dealing with complications, brings unique difficulties. The most striking pathophysiological features of patients with severe COVID-19 are dysregulated immune responses and abnormal coagulation function, which can result in multiple-organ failure and death. Normally metabolized high-density lipoprotein (HDL) performs several functions, including reverse cholesterol transport, direct binding to lipopolysaccharide (LPS) to neutralize LPS activity, regulation of inflammatory response, anti-thrombotic effects, antioxidant, and anti-apoptotic properties. Clinical data shows that significantly decreased HDL levels in patients with COVID-19 are correlated with both disease severity and mortality. However, the role of HDL in COVID-19 and its specific mechanism remain unclear. In this analysis, we review current evidence mainly in the following areas: firstly, the pathophysiological characteristics of COVID-19, secondly, the pleiotropic properties of HDL, thirdly, the changes and clinical significance of HDL in COVID-19, and fourthly the prospect of HDL-targeting therapy in COVID-19 to clarify the role of HDL in the pathogenesis of COVID-19 and discuss the potential of HDL therapy in COVID-19.
Collapse
Affiliation(s)
- Guyi Wang
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jiayi Deng
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jinxiu Li
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chenfang Wu
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Haiyun Dong
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Shangjie Wu
- Department of Respiratory, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yanjun Zhong
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
38
|
Jamaly S, Tsokos MG, Bhargava R, Brook OR, Hecht JL, Abdi R, Moulton VR, Satyam A, Tsokos GC. Complement activation and increased expression of Syk, mucin-1 and CaMK4 in kidneys of patients with COVID-19. Clin Immunol 2021; 229:108795. [PMID: 34252574 PMCID: PMC8270746 DOI: 10.1016/j.clim.2021.108795] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023]
Abstract
Acute and chronic kidney failure is common in hospitalized patients with COVID-19, yet the mechanism of injury and predisposing factors remain poorly understood. We investigated the role of complement activation by determining the levels of deposited complement components (C1q, C3, FH, C5b-9) and immunoglobulin along with the expression levels of the injury-associated molecules spleen tyrosine kinase (Syk), mucin-1 (MUC1) and calcium/calmodulin-dependent protein kinase IV (CaMK4) in the kidney tissues of people who succumbed to COVID-19. We report increased deposition of C1q, C3, C5b-9, total immunoglobulin, and high expression levels of Syk, MUC1 and CaMK4 in the kidneys of COVID-19 patients. Our study provides strong rationale for the expansion of trials involving the use of inhibitors of these molecules, in particular C1q, C3, Syk, MUC1 and CaMK4 to treat patients with COVID-19.
Collapse
Affiliation(s)
- Simin Jamaly
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States; Department of Medical Biology, Faculty of Health Science, UiT Arctic University of Norway, N-9037 Tromsø, Norway
| | - Maria G Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Rhea Bhargava
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Olga R Brook
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Jonathan L Hecht
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Vaishali R Moulton
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Abhigyan Satyam
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States.
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
39
|
Gavriilaki E, Asteris PG, Touloumenidou T, Koravou EE, Koutra M, Papayanni PG, Karali V, Papalexandri A, Varelas C, Chatzopoulou F, Chatzidimitriou M, Chatzidimitriou D, Veleni A, Grigoriadis S, Rapti E, Chloros D, Kioumis I, Kaimakamis E, Bitzani M, Boumpas D, Tsantes A, Sotiropoulos D, Sakellari I, Kalantzis IG, Parastatidis ST, Koopialipoor M, Cavaleri L, Armaghani DJ, Papadopoulou A, Brodsky RA, Kokoris S, Anagnostopoulos A. Genetic justification of severe COVID-19 using a rigorous algorithm. Clin Immunol 2021; 226:108726. [PMID: 33845193 PMCID: PMC8043057 DOI: 10.1016/j.clim.2021.108726] [Citation(s) in RCA: 33] [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] [Received: 03/19/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 01/04/2023]
Abstract
Recent studies suggest excessive complement activation in severe coronavirus disease-19 (COVID-19). The latter shares common characteristics with complement-mediated thrombotic microangiopathy (TMA). We hypothesized that genetic susceptibility would be evident in patients with severe COVID-19 (similar to TMA) and associated with disease severity. We analyzed genetic and clinical data from 97 patients hospitalized for COVID-19. Through targeted next-generation-sequencing we found an ADAMTS13 variant in 49 patients, along with two risk factor variants (C3, 21 patients; CFH,34 patients). 31 (32%) patients had a combination of these, which was independently associated with ICU hospitalization (p = 0.022). Analysis of almost infinite variant combinations showed that patients with rs1042580 in thrombomodulin and without rs800292 in complement factor H did not require ICU hospitalization. We also observed gender differences in ADAMTS13 and complement-related variants. In light of encouraging results by complement inhibitors, our study highlights a patient population that might benefit from early initiation of specific treatment.
Collapse
Affiliation(s)
- Eleni Gavriilaki
- Hematology Department - BMT Unit, G Papanicolaou Hospital, Thessaloniki, Greece.
| | - Panagiotis G Asteris
- Computational Mechanics Laboratory, School of Pedagogical and Technological Education, Athens, Greece
| | | | | | - Maria Koutra
- Hematology Department - BMT Unit, G Papanicolaou Hospital, Thessaloniki, Greece
| | | | - Vassiliki Karali
- Rheumatology and Clinical Immunology Unit, "Attikon" University Hospital, Athens, Greece
| | | | - Christos Varelas
- Hematology Department - BMT Unit, G Papanicolaou Hospital, Thessaloniki, Greece
| | - Fani Chatzopoulou
- Microbiology Department, Aristotle University of Thessaloniki, Greece
| | - Maria Chatzidimitriou
- Biomedical Sciences, Alexander Campus International Hellenic University, Thessaloniki, Greece
| | | | - Anastasia Veleni
- Infectious Disease Committee, G Papanicolaou Hospital, Thessaloniki, Greece
| | - Savvas Grigoriadis
- Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Evdoxia Rapti
- Laboratory of Hematology and Hospital Blood Transfusion Department, University General Hospital "Attikon", NKUA, Medical School, Greece
| | - Diamantis Chloros
- Pneumonology Department, G Papanicolaou Hospital, Thessaloniki, Greece
| | - Ioannis Kioumis
- Respiratory Failure Department, G Papanicolaou Hospital-Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Milly Bitzani
- 1st Intensive Care Unit, G Papanicolaou Hospital, Thessaloniki, Greece
| | - Dimitrios Boumpas
- Rheumatology and Clinical Immunology Unit, "Attikon" University Hospital, Athens, Greece
| | - Argyris Tsantes
- Laboratory of Hematology and Hospital Blood Transfusion Department, University General Hospital "Attikon", NKUA, Medical School, Greece
| | | | - Ioanna Sakellari
- Hematology Department - BMT Unit, G Papanicolaou Hospital, Thessaloniki, Greece
| | | | - Stefanos T Parastatidis
- Computational Mechanics Laboratory, School of Pedagogical and Technological Education, Athens, Greece
| | | | - Liborio Cavaleri
- Department of Civil, Environmental, Aerospace and Materials Engineering, University of Palermo, Palermo, Italy
| | - Danial J Armaghani
- Department of Civil Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
| | | | - Robert Alan Brodsky
- Hematology Division, Department of Internal Medicine, Johns Hopkins University, Baltimore, USA
| | - Styliani Kokoris
- Laboratory of Hematology and Hospital Blood Transfusion Department, University General Hospital "Attikon", NKUA, Medical School, Greece
| | | |
Collapse
|