1
|
Monarca L, Ragonese F, Biagini A, Sabbatini P, Pacini M, Zucchi A, Spaccapelo R, Ferrari P, Nicolini A, Fioretti B. Electrophysiological Impact of SARS-CoV-2 Envelope Protein in U251 Human Glioblastoma Cells: Possible Implications in Gliomagenesis? Int J Mol Sci 2024; 25:6669. [PMID: 38928376 PMCID: PMC11203726 DOI: 10.3390/ijms25126669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/28/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
SARS-CoV-2 is the causative agent of the COVID-19 pandemic, the acute respiratory disease which, so far, has led to over 7 million deaths. There are several symptoms associated with SARS-CoV-2 infections which include neurological and psychiatric disorders, at least in the case of pre-Omicron variants. SARS-CoV-2 infection can also promote the onset of glioblastoma in patients without prior malignancies. In this study, we focused on the Envelope protein codified by the virus genome, which acts as viroporin and that is reported to be central for virus propagation. In particular, we characterized the electrophysiological profile of E-protein transfected U251 and HEK293 cells through the patch-clamp technique and FURA-2 measurements. Specifically, we observed an increase in the voltage-dependent (Kv) and calcium-dependent (KCa) potassium currents in HEK293 and U251 cell lines, respectively. Interestingly, in both cellular models, we observed a depolarization of the mitochondrial membrane potential in accordance with an alteration of U251 cell growth. We, therefore, investigated the transcriptional effect of E protein on the signaling pathways and found several gene alterations associated with apoptosis, cytokines and WNT pathways. The electrophysiological and transcriptional changes observed after E protein expression could explain the impact of SARS-CoV-2 infection on gliomagenesis.
Collapse
Affiliation(s)
- Lorenzo Monarca
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, 06123 Perugia, Italy; (L.M.); (F.R.); (A.B.); (P.S.)
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, 06132 Perugia, Italy;
| | - Francesco Ragonese
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, 06123 Perugia, Italy; (L.M.); (F.R.); (A.B.); (P.S.)
| | - Andrea Biagini
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, 06123 Perugia, Italy; (L.M.); (F.R.); (A.B.); (P.S.)
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, 06132 Perugia, Italy;
| | - Paola Sabbatini
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, 06123 Perugia, Italy; (L.M.); (F.R.); (A.B.); (P.S.)
| | - Matteo Pacini
- Urology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (M.P.); (A.Z.)
| | - Alessandro Zucchi
- Urology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (M.P.); (A.Z.)
| | - Roberta Spaccapelo
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, 06132 Perugia, Italy;
- Interuniversity Consortium for Biotechnology (C.I.B.), 34148 Trieste, Italy
| | - Paola Ferrari
- Department of Oncology, Transplantations and New Technologies in Medicine, University of Pisa, 56126 Pisa, Italy;
| | - Andrea Nicolini
- Department of Oncology, Transplantations and New Technologies in Medicine, University of Pisa, 56126 Pisa, Italy;
| | - Bernard Fioretti
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, 06123 Perugia, Italy; (L.M.); (F.R.); (A.B.); (P.S.)
| |
Collapse
|
2
|
Gajate-Arenas M, Fricke-Galindo I, García-Pérez O, Domínguez-de-Barros A, Pérez-Rubio G, Dorta-Guerra R, Buendía-Roldán I, Chávez-Galán L, Lorenzo-Morales J, Falfán-Valencia R, Córdoba-Lanús E. The Immune Response of OAS1, IRF9, and IFI6 Genes in the Pathogenesis of COVID-19. Int J Mol Sci 2024; 25:4632. [PMID: 38731851 PMCID: PMC11083791 DOI: 10.3390/ijms25094632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
COVID-19 is characterized by a wide range of clinical manifestations, where aging, underlying diseases, and genetic background are related to worse outcomes. In the present study, the differential expression of seven genes related to immunity, IRF9, CCL5, IFI6, TGFB1, IL1B, OAS1, and TFRC, was analyzed in individuals with COVID-19 diagnoses of different disease severities. Two-step RT-qPCR was performed to determine the relative gene expression in whole-blood samples from 160 individuals. The expression of OAS1 (p < 0.05) and IFI6 (p < 0.05) was higher in moderate hospitalized cases than in severe ones. Increased gene expression of OAS1 (OR = 0.64, CI = 0.52-0.79; p = 0.001), IRF9 (OR = 0.581, CI = 0.43-0.79; p = 0.001), and IFI6 (OR = 0.544, CI = 0.39-0.69; p < 0.001) was associated with a lower risk of requiring IMV. Moreover, TGFB1 (OR = 0.646, CI = 0.50-0.83; p = 0.001), CCL5 (OR = 0.57, CI = 0.39-0.83; p = 0.003), IRF9 (OR = 0.80, CI = 0.653-0.979; p = 0.03), and IFI6 (OR = 0.827, CI = 0.69-0.991; p = 0.039) expression was associated with patient survival. In conclusion, the relevance of OAS1, IRF9, and IFI6 in controlling the viral infection was confirmed.
Collapse
Affiliation(s)
- Malena Gajate-Arenas
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, 38029 San Cristóbal de La Laguna, Spain; (M.G.-A.); (O.G.-P.); (A.D.-d.-B.); (R.D.-G.)
| | - Ingrid Fricke-Galindo
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.F.-G.); (G.P.-R.); (R.F.-V.)
| | - Omar García-Pérez
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, 38029 San Cristóbal de La Laguna, Spain; (M.G.-A.); (O.G.-P.); (A.D.-d.-B.); (R.D.-G.)
| | - Angélica Domínguez-de-Barros
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, 38029 San Cristóbal de La Laguna, Spain; (M.G.-A.); (O.G.-P.); (A.D.-d.-B.); (R.D.-G.)
| | - Gloria Pérez-Rubio
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.F.-G.); (G.P.-R.); (R.F.-V.)
| | - Roberto Dorta-Guerra
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, 38029 San Cristóbal de La Laguna, Spain; (M.G.-A.); (O.G.-P.); (A.D.-d.-B.); (R.D.-G.)
- Department of Mathematics, Statistics and Operations Research, Faculty of Sciences, Mathematics Section, Universidad de La Laguna, 38200 San Cristóbal de La Laguna, Spain
| | - Ivette Buendía-Roldán
- Translational Research Laboratory on Aging and Pulmonary Fibrosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico;
| | - Leslie Chávez-Galán
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City 14080, Mexico;
| | - Jacob Lorenzo-Morales
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, 38029 San Cristóbal de La Laguna, Spain; (M.G.-A.); (O.G.-P.); (A.D.-d.-B.); (R.D.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Obstetrics and Gynecology, Pediatrics, Preventive Medicine and Public Health, Toxicology, Legal and Forensic Medicine and Parasitology, Faculty of Health Sciences, Universidad de La Laguna, 38200 San Cristóbal de La Laguna, Spain
| | - Ramcés Falfán-Valencia
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.F.-G.); (G.P.-R.); (R.F.-V.)
| | - Elizabeth Córdoba-Lanús
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, 38029 San Cristóbal de La Laguna, Spain; (M.G.-A.); (O.G.-P.); (A.D.-d.-B.); (R.D.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
3
|
Ferreira JM, dos Santos BRC, de Moura EL, dos Santos ACM, Vencioneck Dutra JC, Figueiredo EVMDS, de Lima Filho JL. Narrowing the Relationship between Human CCR5 Gene Polymorphisms and Chagas Disease: Systematic Review and Meta-Analysis. Life (Basel) 2023; 13:1677. [PMID: 37629534 PMCID: PMC10455882 DOI: 10.3390/life13081677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
Our aim was to carry out a qualitative and quantitative synthesis of the influence of CCR5 genetic variants on Chagas disease (CD) through a systematic review. A total of 1197 articles were analyzed, and eleven were included in the review. A meta-analysis was conducted along with principal component analyses (PCAs). The polymorphisms found were analyzed using the SNP2TFBS tool to identify possible variants that influence the interaction with gene binding sites. Eleven studied variants were identified: rs2856758, rs2734648, rs1799987, rs1799988, rs41469351, rs1800023, rs1800024, Δ32/rs333, rs3176763, rs3087253 and rs11575815. The studies analyzed were published between 2001 and 2019, conducted in Argentina, Brazil, Spain, Colombia and Venezuela, and included Argentine, Brazilian, Colombian, Peruvian and Venezuelan patients. Eight polymorphisms were subjected to the meta-analysis, of which six were associated with the development of the cardiac form of CD: rs1799987-G/G and G/A in the dominance model and G/G in the recessiveness model; rs2856758-A/G in the codominance model; rs2734648-T/T and T/G in the dominance model; rs1799988-T/T in both the codominance and recessiveness models; rs1800023-G allele and the G/G genotype in the codominance and recessiveness models, and the G/G and G/A genotypes in the dominance model; and rs1800024-T allele. The PCA analyses were able to indicate the relationships between the alleles and the genotypes of the polymorphisms. The SNP2TFBS tool identified rs1800023 as an influencer of the Spi1 transcription factor (p < 0.05). A correlation was established between the alleles associated with the cardiac form of CD in this review, members of the C haplotype of the gene (HHC-TGTG), and the cardiac form of CD.
Collapse
Affiliation(s)
- Jean Moisés Ferreira
- Laboratório de Imunopatologia Keizo Asami—LIKA, Centro de Biocièncias, Universidade Federal de Pernambuco (UFPE), Recife 50670-901, Pernambuco, Brazil
- Secretaria de Estado de Educação do Espírito Santo (SEDU), Santa Lucia, Vitória 29056-085, Espírito Santo, Brazil;
| | - Barbara Rayssa Correia dos Santos
- Laboratório de Biologia Molecular e Expressão Gênica—LABMEG, Departamento de Ciências Biológicas, Universidade Federal de Alagoas (UFAL), Campus Arapiraca, Arapiraca 57300-970, Alagoas, Brazil; (B.R.C.d.S.); (E.L.d.M.); (A.C.M.d.S.); (E.V.M.d.S.F.)
| | - Edilson Leite de Moura
- Laboratório de Biologia Molecular e Expressão Gênica—LABMEG, Departamento de Ciências Biológicas, Universidade Federal de Alagoas (UFAL), Campus Arapiraca, Arapiraca 57300-970, Alagoas, Brazil; (B.R.C.d.S.); (E.L.d.M.); (A.C.M.d.S.); (E.V.M.d.S.F.)
| | - Ana Caroline Melo dos Santos
- Laboratório de Biologia Molecular e Expressão Gênica—LABMEG, Departamento de Ciências Biológicas, Universidade Federal de Alagoas (UFAL), Campus Arapiraca, Arapiraca 57300-970, Alagoas, Brazil; (B.R.C.d.S.); (E.L.d.M.); (A.C.M.d.S.); (E.V.M.d.S.F.)
| | - Jean Carlos Vencioneck Dutra
- Secretaria de Estado de Educação do Espírito Santo (SEDU), Santa Lucia, Vitória 29056-085, Espírito Santo, Brazil;
| | - Elaine Virgínia Martins de Sousa Figueiredo
- Laboratório de Biologia Molecular e Expressão Gênica—LABMEG, Departamento de Ciências Biológicas, Universidade Federal de Alagoas (UFAL), Campus Arapiraca, Arapiraca 57300-970, Alagoas, Brazil; (B.R.C.d.S.); (E.L.d.M.); (A.C.M.d.S.); (E.V.M.d.S.F.)
| | - José Luiz de Lima Filho
- Laboratório de Imunopatologia Keizo Asami—LIKA, Centro de Biocièncias, Universidade Federal de Pernambuco (UFPE), Recife 50670-901, Pernambuco, Brazil
| |
Collapse
|
4
|
Mali A, Franci G, Zannella C, Chianese A, Anthiya S, López-Estévez AM, Monti A, De Filippis A, Doti N, Alonso MJ, Galdiero M. Antiviral Peptides Delivered by Chitosan-Based Nanoparticles to Neutralize SARS-CoV-2 and HCoV-OC43. Pharmaceutics 2023; 15:1621. [PMID: 37376070 DOI: 10.3390/pharmaceutics15061621] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/29/2023] [Accepted: 04/07/2023] [Indexed: 06/29/2023] Open
Abstract
The COVID-19 pandemic has made it clear that there is a crucial need for the design and development of antiviral agents that can efficiently reduce the fatality rate caused by infectious diseases. The fact that coronavirus mainly enters through the nasal epithelial cells and spreads through the nasal passage makes the nasal delivery of antiviral agents a promising strategy not only to reduce viral infection but also its transmission. Peptides are emerging as powerful candidates for antiviral treatments, showing not only a strong antiviral activity, but also improved safety, efficacy, and higher specificity against viral pathogens. Based on our previous experience on the use of chitosan-based nanoparticles to deliver peptides intra-nasally the current study aimed to explore the delivery of two-novel antiviral peptides making use of nanoparticles consisting of HA/CS and DS/CS. The antiviral peptides were chemically synthesized, and the optimal conditions for encapsulating them were selected through a combination of physical entrapment and chemical conjugation using HA/CS and DS/CS nanocomplexes. Finally, we evaluated the in vitro neutralization capacity against SARS-CoV-2 and HCoV-OC43 for potential use as prophylaxis or therapy.
Collapse
Affiliation(s)
- Avinash Mali
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Gianluigi Franci
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana", University of Salerno, 84081 Baronissi, Italy
| | - Carla Zannella
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Annalisa Chianese
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Shubaash Anthiya
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute of Santiago de Compostela (IDIS), Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Ana M López-Estévez
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute of Santiago de Compostela (IDIS), Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Alessandra Monti
- Institute of Biostructures and Bioimaging (IBB), National Research Council (CNR), 80131 Naples, Italy
- CIRPEB, Centro Interuniversitario di Ricerca sui Peptidi Bioattivi, 80134 Naples, Italy
| | - Anna De Filippis
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Nunzianna Doti
- Institute of Biostructures and Bioimaging (IBB), National Research Council (CNR), 80131 Naples, Italy
- CIRPEB, Centro Interuniversitario di Ricerca sui Peptidi Bioattivi, 80134 Naples, Italy
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute of Santiago de Compostela (IDIS), Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Massimiliano Galdiero
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| |
Collapse
|
5
|
SARS-CoV-2 N protein mediates intercellular nucleic acid dispersion, a feature reduced in Omicron. iScience 2023; 26:105995. [PMID: 36687314 PMCID: PMC9841735 DOI: 10.1016/j.isci.2023.105995] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/21/2022] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
The coronavirus nucleocapsid (N) protein is known to bind to nucleic acids and facilitate viral genome encapsulation. Here we report that the N protein can mediate RNA or DNA entering neighboring cells through ACE2-independent, receptor (STEAP2)-mediated endocytosis, and achieve gene expression. The effect is more pronounced for the N protein of wild-type SARS-CoV-2 than that of the Omicron variant and other human coronaviruses. This effect is enhanced by RANTES (CCL5), a chemokine induced by N protein, and lactate, a metabolite produced in hypoxia, to cause more damage. These findings might explain the clinical observations in SARS-CoV-2-infected cases. Moreover, the N protein-mediated function can be inhibited by N protein-specific monoclonal antibodies or p38 mitogen-activated protein kinase inhibitors. Since the N-protein-mediated nucleic acid endocytosis involves a receptor commonly expressed in many types of cells, our findings suggest that N protein may have an additional role in SARS-CoV-2 pathogenesis.
Collapse
|
6
|
Fields AT, Andraska EA, Kaltenmeier C, Matthay ZA, Herrera K, Nuñez-Garcia B, Jones CM, Wick KD, Liu S, Luo JH, Yu YP, Matthay MA, Hendrickson CM, Bainton RJ, Barrett TJ, Berger JS, Neal MD, Kornblith LZ. Effects of the circulating environment of COVID-19 on platelet and neutrophil behavior. Front Immunol 2023; 14:1130288. [PMID: 36999030 PMCID: PMC10043426 DOI: 10.3389/fimmu.2023.1130288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/23/2023] [Indexed: 04/01/2023] Open
Abstract
Introduction Thromboinflammatory complications are well described sequalae of Coronavirus Disease 2019 (COVID-19), and there is evidence of both hyperreactive platelet and inflammatory neutrophil biology that contributes to the thromoinflammatory milieu. It has been demonstrated in other thromboinflammatory diseases that the circulating environment may affect cellular behavior, but what role this environment exerts on platelets and neutrophils in COVID-19 remains unknown. We tested the hypotheses that 1) plasma from COVID-19 patients can induce a prothrombotic platelet functional phenotype, and 2) contents released from platelets (platelet releasate) from COVID-19 patients can induce a proinflammatory neutrophil phenotype. Methods We treated platelets with COVID-19 patient and disease control plasma, and measured their aggregation response to collagen and adhesion in a microfluidic parallel plate flow chamber coated with collagen and thromboplastin. We exposed healthy neutrophils to platelet releasate from COVID-19 patients and disease controls and measured neutrophil extracellular trap formation and performed RNA sequencing. Results We found that COVID-19 patient plasma promoted auto-aggregation, thereby reducing response to further stimulation ex-vivo. Neither disease condition increased the number of platelets adhered to a collagen and thromboplastin coated parallel plate flow chamber, but both markedly reduced platelet size. COVID-19 patient platelet releasate increased myeloperoxidasedeoxyribonucleic acid complexes and induced changes to neutrophil gene expression. Discussion Together these results suggest aspects of the soluble environment circulating platelets, and that the contents released from those neutrophil behavior independent of direct cellular contact.
Collapse
Affiliation(s)
- Alexander T. Fields
- Department of Surgery, University of California, San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, United States
| | - Elizabeth A. Andraska
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Christof Kaltenmeier
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Zachary A. Matthay
- Department of Surgery, University of California, San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, United States
| | - Kimberly Herrera
- Department of Surgery, University of California, San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, United States
| | - Brenda Nuñez-Garcia
- Department of Surgery, University of California, San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, United States
| | - Chayse M. Jones
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Katherine D. Wick
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Silvia Liu
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jian-Hua Luo
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Yan-Ping Yu
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michael A. Matthay
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Carolyn M. Hendrickson
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Roland J. Bainton
- Department of Anesthesia and Perioperative Care, School of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Tessa J. Barrett
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University (NYU) Grossman School of Medicine, New York, NY, United States
| | - Jeffrey S. Berger
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University (NYU) Grossman School of Medicine, New York, NY, United States
- New York University (NYU) Center for the Prevention of Cardiovascular Disease, New York University (NYU) Langone Health, New York, NY, United States
- Division of Vascular Surgery, Department of Surgery, New York University (NYU) Grossman School of Medicine, New York, NY, United States
| | - Matthew D. Neal
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Lucy Z. Kornblith
- Department of Surgery, University of California, San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, United States
- *Correspondence: Lucy Z. Kornblith,
| |
Collapse
|
7
|
Hönzke K, Obermayer B, Mache C, Fatykhova D, Kessler M, Dökel S, Wyler E, Baumgardt M, Löwa A, Hoffmann K, Graff P, Schulze J, Mieth M, Hellwig K, Demir Z, Biere B, Brunotte L, Mecate-Zambrano A, Bushe J, Dohmen M, Hinze C, Elezkurtaj S, Tönnies M, Bauer TT, Eggeling S, Tran HL, Schneider P, Neudecker J, Rückert JC, Schmidt-Ott KM, Busch J, Klauschen F, Horst D, Radbruch H, Radke J, Heppner F, Corman VM, Niemeyer D, Müller MA, Goffinet C, Mothes R, Pascual-Reguant A, Hauser AE, Beule D, Landthaler M, Ludwig S, Suttorp N, Witzenrath M, Gruber AD, Drosten C, Sander LE, Wolff T, Hippenstiel S, Hocke AC. Human lungs show limited permissiveness for SARS-CoV-2 due to scarce ACE2 levels but virus-induced expansion of inflammatory macrophages. Eur Respir J 2022; 60:2102725. [PMID: 35728978 PMCID: PMC9712848 DOI: 10.1183/13993003.02725-2021] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 05/25/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) utilises the angiotensin-converting enzyme 2 (ACE2) transmembrane peptidase as cellular entry receptor. However, whether SARS-CoV-2 in the alveolar compartment is strictly ACE2-dependent and to what extent virus-induced tissue damage and/or direct immune activation determines early pathogenesis is still elusive. METHODS Spectral microscopy, single-cell/-nucleus RNA sequencing or ACE2 "gain-of-function" experiments were applied to infected human lung explants and adult stem cell derived human lung organoids to correlate ACE2 and related host factors with SARS-CoV-2 tropism, propagation, virulence and immune activation compared to SARS-CoV, influenza and Middle East respiratory syndrome coronavirus (MERS-CoV). Coronavirus disease 2019 (COVID-19) autopsy material was used to validate ex vivo results. RESULTS We provide evidence that alveolar ACE2 expression must be considered scarce, thereby limiting SARS-CoV-2 propagation and virus-induced tissue damage in the human alveolus. Instead, ex vivo infected human lungs and COVID-19 autopsy samples showed that alveolar macrophages were frequently positive for SARS-CoV-2. Single-cell/-nucleus transcriptomics further revealed nonproductive virus uptake and a related inflammatory and anti-viral activation, especially in "inflammatory alveolar macrophages", comparable to those induced by SARS-CoV and MERS-CoV, but different from NL63 or influenza virus infection. CONCLUSIONS Collectively, our findings indicate that severe lung injury in COVID-19 probably results from a macrophage-triggered immune activation rather than direct viral damage of the alveolar compartment.
Collapse
Affiliation(s)
- Katja Hönzke
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Contributed equally
| | - Benedikt Obermayer
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Core Unit Bioinformatics, Berlin, Germany
- Contributed equally
| | - Christin Mache
- Unit 17 "Influenza and other Respiratory Viruses", Robert Koch Institut, Berlin, Germany
- Contributed equally
| | - Diana Fatykhova
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mirjana Kessler
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Gynecology and Obstetrics, Ludwig-Maximilian University, Munich, Germany
| | - Simon Dökel
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Emanuel Wyler
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) and IRI Life Sciences, Institute for Biology, Humboldt Universität zu Berlin, Berlin, Germany
| | - Morris Baumgardt
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Anna Löwa
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Karen Hoffmann
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Patrick Graff
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jessica Schulze
- Unit 17 "Influenza and other Respiratory Viruses", Robert Koch Institut, Berlin, Germany
| | - Maren Mieth
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Katharina Hellwig
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Zeynep Demir
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Barbara Biere
- Unit 17 "Influenza and other Respiratory Viruses", Robert Koch Institut, Berlin, Germany
| | - Linda Brunotte
- Institute of Virology, Westfaelische Wilhelms Universität, Münster, Germany
| | | | - Judith Bushe
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Melanie Dohmen
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Christian Hinze
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sefer Elezkurtaj
- Department of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mario Tönnies
- HELIOS Clinic Emil von Behring, Department of Pneumology and Department of Thoracic Surgery, Chest Hospital Heckeshorn, Berlin, Germany
| | - Torsten T Bauer
- HELIOS Clinic Emil von Behring, Department of Pneumology and Department of Thoracic Surgery, Chest Hospital Heckeshorn, Berlin, Germany
| | - Stephan Eggeling
- Department of Thoracic Surgery, Vivantes Clinics Neukölln, Berlin, Germany
| | - Hong-Linh Tran
- Department of Thoracic Surgery, Vivantes Clinics Neukölln, Berlin, Germany
| | - Paul Schneider
- Department for Thoracic Surgery, DRK Clinics, Berlin, Germany
| | - Jens Neudecker
- Department of General, Visceral, Vascular and Thoracic Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jens C Rückert
- Department of General, Visceral, Vascular and Thoracic Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kai M Schmidt-Ott
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jonas Busch
- Clinic for Urology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Frederick Klauschen
- Department of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - David Horst
- Department of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Helena Radbruch
- Institute for Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Josefine Radke
- Institute for Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Frank Heppner
- Institute for Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Victor M Corman
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Daniela Niemeyer
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Marcel A Müller
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christine Goffinet
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ronja Mothes
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Anna Pascual-Reguant
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Anja Erika Hauser
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Dieter Beule
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Core Unit Bioinformatics, Berlin, Germany
| | - Markus Landthaler
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC) and IRI Life Sciences, Institute for Biology, Humboldt Universität zu Berlin, Berlin, Germany
| | - Stephan Ludwig
- Institute of Virology, Westfaelische Wilhelms Universität, Münster, 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
| | - Martin Witzenrath
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Achim D Gruber
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Christian Drosten
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Leif-Erik Sander
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Thorsten Wolff
- Unit 17 "Influenza and other Respiratory Viruses", Robert Koch Institut, 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
| | - Andreas C Hocke
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
8
|
Fischer JC, Balz V, Jazmati D, Bölke E, Freise NF, Keitel V, Feldt T, Jensen BEO, Bode J, Lüdde T, Häussinger D, Adams O, Schneider EM, Enczmann J, Rox JM, Hermsen D, Schulze-Bosse K, Kindgen-Milles D, Knoefel WT, van Griensven M, Haussmann J, Tamaskovics B, Plettenberg C, Scheckenbach K, Corradini S, Pedoto A, Maas K, Schmidt L, Grebe O, Esposito I, Ehrhardt A, Peiper M, Buhren BA, Calles C, Stöhr A, Gerber PA, Lichtenberg A, Schelzig H, Flaig Y, Rezazadeh A, Budach W, Matuschek C. Prognostic markers for the clinical course in the blood of patients with SARS-CoV-2 infection. Eur J Med Res 2022; 27:255. [PMID: 36411478 PMCID: PMC9676819 DOI: 10.1186/s40001-022-00864-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/20/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND The presentation of peptides and the subsequent immune response depend on the MHC characteristics and influence the specificity of the immune response. Several studies have found an association between HLA variants and differential COVID-19 outcomes and have shown that HLA genotypes are associated with differential immune responses against SARS-CoV-2, particularly in severely ill patients. Information, whether HLA haplotypes are associated with the severity or length of the disease in moderately diseased individuals is absent. METHODS Next-generation sequencing-based HLA typing was performed in 303 female and 231 male non-hospitalized North Rhine Westphalian patients infected with SARS-CoV2 during the first and second wave. For HLA-Class I, we obtained results from 528 patients, and for HLA-Class II from 531. In those patients, who became ill between March 2020 and January 2021, the 22 most common HLA-Class I (HLA-A, -B, -C) or HLA-Class II (HLA -DRB1/3/4, -DQA1, -DQB1) haplotypes were determined. The identified HLA haplotypes as well as the presence of a CCR5Δ32 mutation and number of O and A blood group alleles were associated to disease severity and duration of the disease. RESULTS The influence of the HLA haplotypes on disease severity and duration was more pronounced than the influence of age, sex, or ABO blood group. These associations were sex dependent. The presence of mutated CCR5 resulted in a longer recovery period in males. CONCLUSION The existence of certain HLA haplotypes is associated with more severe disease.
Collapse
Affiliation(s)
- Johannes C. Fischer
- grid.14778.3d0000 0000 8922 7789Institute for Transplant Diagnostics and Cell Therapeutics, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Vera Balz
- grid.14778.3d0000 0000 8922 7789Institute for Transplant Diagnostics and Cell Therapeutics, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Danny Jazmati
- grid.14778.3d0000 0000 8922 7789Department of Radiation Oncology, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Edwin Bölke
- grid.14778.3d0000 0000 8922 7789Department of Radiation Oncology, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Noemi F. Freise
- grid.14778.3d0000 0000 8922 7789Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Verena Keitel
- grid.14778.3d0000 0000 8922 7789Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Torsten Feldt
- grid.14778.3d0000 0000 8922 7789Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Björn-Erik Ole Jensen
- grid.14778.3d0000 0000 8922 7789Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Johannes Bode
- grid.14778.3d0000 0000 8922 7789Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Tom Lüdde
- grid.14778.3d0000 0000 8922 7789Institute for Virology, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Dieter Häussinger
- grid.14778.3d0000 0000 8922 7789Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Ortwin Adams
- grid.14778.3d0000 0000 8922 7789Institute for Virology, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - E. Marion Schneider
- grid.410712.10000 0004 0473 882XDivision of Experimental Anesthesiology, University Hospital Ulm, Ulm, Germany
| | - Jürgen Enczmann
- grid.14778.3d0000 0000 8922 7789Institute for Transplant Diagnostics and Cell Therapeutics, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Jutta M. Rox
- grid.14778.3d0000 0000 8922 7789Institute for Transplant Diagnostics and Cell Therapeutics, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Derik Hermsen
- grid.14778.3d0000 0000 8922 7789Central Institute for Laboratory Diagnostics and Clinical Chemistry, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Karin Schulze-Bosse
- grid.14778.3d0000 0000 8922 7789Central Institute for Laboratory Diagnostics and Clinical Chemistry, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Detlef Kindgen-Milles
- grid.14778.3d0000 0000 8922 7789Department of Anesthesiology, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Wolfram Trudo Knoefel
- grid.14778.3d0000 0000 8922 7789Department of Surgery and Interdisciplinary Surgical Intensive Care Unit, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Martijn van Griensven
- grid.5012.60000 0001 0481 6099Department cBITE, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Jan Haussmann
- grid.14778.3d0000 0000 8922 7789Department of Radiation Oncology, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Balint Tamaskovics
- grid.14778.3d0000 0000 8922 7789Department of Radiation Oncology, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Christian Plettenberg
- grid.14778.3d0000 0000 8922 7789Department of Ear, Nose and Throat Disease, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Kathrin Scheckenbach
- grid.14778.3d0000 0000 8922 7789Department of Ear, Nose and Throat Disease, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Stefanie Corradini
- grid.5252.00000 0004 1936 973XDepartment of Radiation Oncology, LMU University of Munich, Munich, Germany
| | - Alessia Pedoto
- grid.51462.340000 0001 2171 9952Department of Anesthesiology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Kitti Maas
- grid.14778.3d0000 0000 8922 7789Department of Radiation Oncology, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Livia Schmidt
- grid.14778.3d0000 0000 8922 7789Department of Radiation Oncology, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Olaf Grebe
- Department of Cardiology and Rhythmology, Petrus Hospital, Wuppertal, Germany
| | - Irene Esposito
- grid.14778.3d0000 0000 8922 7789Institute of Pathology, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Anja Ehrhardt
- grid.412581.b0000 0000 9024 6397Institute of Virology, University of Witten/Herdecke, Witten, Germany
| | - Matthias Peiper
- grid.14778.3d0000 0000 8922 7789Medical Faculty, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Bettina Alexandra Buhren
- grid.14778.3d0000 0000 8922 7789Department of Radiation Oncology, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Christian Calles
- grid.14778.3d0000 0000 8922 7789Coordination Center for Clinical Studies, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Andreas Stöhr
- grid.14778.3d0000 0000 8922 7789Coordination Center for Clinical Studies, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Peter Arne Gerber
- grid.14778.3d0000 0000 8922 7789Department of Radiation Oncology, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Artur Lichtenberg
- grid.14778.3d0000 0000 8922 7789Department of Cardiac Surgery, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Hubert Schelzig
- grid.14778.3d0000 0000 8922 7789Department of Vascular Surgery, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Yechan Flaig
- grid.14778.3d0000 0000 8922 7789Department of Radiation Oncology, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Amir Rezazadeh
- grid.14778.3d0000 0000 8922 7789Department of Radiation Oncology, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Wilfried Budach
- grid.14778.3d0000 0000 8922 7789Department of Radiation Oncology, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| | - Christiane Matuschek
- grid.14778.3d0000 0000 8922 7789Department of Radiation Oncology, Medical Faculty, University Hospital Dusseldorf, Heinrich-Heine-University Dusseldorf, Dusseldorf, Germany
| |
Collapse
|
9
|
Karimabad MN, Hassanshahi G, Kounis NG, Mplani V, Roditis P, Gogos C, Lagadinou M, Assimakopoulos SF, Dousdampanis P, Koniari I. The Chemokines CXC, CC and C in the Pathogenesis of COVID-19 Disease and as Surrogates of Vaccine-Induced Innate and Adaptive Protective Responses. Vaccines (Basel) 2022; 10:vaccines10081299. [PMID: 36016187 PMCID: PMC9416781 DOI: 10.3390/vaccines10081299] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/08/2022] [Accepted: 08/08/2022] [Indexed: 02/07/2023] Open
Abstract
COVID-19 is one of the progressive viral pandemics that originated from East Asia. COVID-19 or SARS-CoV-2 has been shown to be associated with a chain of physio-pathological mechanisms that are basically immunological in nature. In addition, chemokines have been proposed as a subgroup of chemotactic cytokines with different activities ranging from leukocyte recruitment to injury sites, irritation, and inflammation to angiostasis and angiogenesis. Therefore, researchers have categorized the chemotactic elements into four classes, including CX3C, CXC, CC, and C, based on the location of the cysteine motifs in their structures. Considering the severe cases of COVID-19, the hyperproduction of particular chemokines occurring in lung tissue as well as pro-inflammatory cytokines significantly worsen the disease prognosis. According to the studies conducted in the field documenting the changing expression of CXC and CC chemokines in COVID-19 cases, the CC and CXC chemokines contribute to this pandemic, and their impact could reflect the development of reasonable strategies for COVID-19 management. The CC and the CXC families of chemokines are important in host immunity to viral infections and along with other biomarkers can serve as the surrogates of vaccine-induced innate and adaptive protective responses, facilitating the improvement of vaccine efficacy. Furthermore, the immunogenicity elicited by the chemokine response to adenovirus vector vaccines may constitute the basis of vaccine-induced immune thrombotic thrombocytopaenia.
Collapse
Affiliation(s)
- Mojgan Noroozi Karimabad
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan 7717933777, Iran
| | - Gholamhossein Hassanshahi
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan 7717933777, Iran
| | - Nicholas G. Kounis
- Department of Internal Medicine, Division of Cardiology, University of Patras Medical School, 26500 Patras, Greece
- Correspondence:
| | - Virginia Mplani
- Intensive Care Unit, Patras University Hospital, 26500 Patras, Greece
| | - Pavlos Roditis
- Department of Cardiology, Mamatsio Kozanis General Hospital, 50100 Kozani, Greece
| | - Christos Gogos
- COVID-19 Unit, Papageorgiou General Hospital, 56403 Thessaloniki, Greece
| | - Maria Lagadinou
- Department of Internal Medicine, Division of Infectious Diseases, University of Patras Medical School, 26500 Patras, Greece
| | - Stelios F. Assimakopoulos
- Department of Internal Medicine, Division of Infectious Diseases, University of Patras Medical School, 26500 Patras, Greece
| | - Periklis Dousdampanis
- Department of Nephrology, Saint Andrews State General Hospital, 26221 Patras, Greece
| | - Ioanna Koniari
- Department of Cardiology, University Hospital of South Manchester, NHS Foundation Trust, Manchester M23 9LT, UK
| |
Collapse
|
10
|
Pacheco-Hernández LM, Ramírez-Noyola JA, Gómez-García IA, Ignacio-Cortés S, Zúñiga J, Choreño-Parra JA. Comparing the Cytokine Storms of COVID-19 and Pandemic Influenza. J Interferon Cytokine Res 2022; 42:369-392. [PMID: 35674675 PMCID: PMC9422807 DOI: 10.1089/jir.2022.0029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 04/19/2022] [Indexed: 12/15/2022] Open
Abstract
Emerging respiratory viruses are major health threats due to their potential to cause massive outbreaks. Over the past 2 years, the coronavirus disease 2019 (COVID-19) pandemic has caused millions of cases of severe infection and deaths worldwide. Although natural and vaccine-induced protective immune mechanisms against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have been increasingly identified, the factors that determine morbimortality are less clear. Comparing the immune signatures of COVID-19 and other severe respiratory infections such as the pandemic influenza might help dissipate current controversies about the origin of their severe manifestations. As such, identifying homologies in the immunopathology of both diseases could provide targets for immunotherapy directed to block shared pathogenic mechanisms. Meanwhile, finding unique characteristics that differentiate each infection could shed light on specific immune alterations exploitable for diagnostic and individualized therapeutics for each case. In this study, we summarize immunopathological aspects of COVID-19 and pandemic influenza from the perspective of cytokine storms as the driving force underlying morbidity. Thereby, we analyze similarities and differences in the cytokine profiles of both infections, aiming to bring forward those molecules more attractive for translational medicine and drug development.
Collapse
Affiliation(s)
- Lynette Miroslava Pacheco-Hernández
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas,” Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Jazmín Ariadna Ramírez-Noyola
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas,” Mexico City, Mexico
- Programa de Maestría en Ciencias de la Salud, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Salvador Díaz Mirón and Plan de San Luis, Mexico City, Mexico
| | - Itzel Alejandra Gómez-García
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas,” Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Sergio Ignacio-Cortés
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas,” Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Joaquín Zúñiga
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas,” Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - José Alberto Choreño-Parra
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas,” Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| |
Collapse
|
11
|
Beirag N, Kumar C, Madan T, Shamji MH, Bulla R, Mitchell D, Murugaiah V, Neto MM, Temperton N, Idicula-Thomas S, Varghese PM, Kishore U. Human surfactant protein D facilitates SARS-CoV-2 pseudotype binding and entry in DC-SIGN expressing cells, and downregulates spike protein induced inflammation. Front Immunol 2022; 13:960733. [PMID: 35967323 PMCID: PMC9367475 DOI: 10.3389/fimmu.2022.960733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Lung surfactant protein D (SP-D) and Dendritic cell-specific intercellular adhesion molecules-3 grabbing non-integrin (DC-SIGN) are pathogen recognising C-type lectin receptors. SP-D has a crucial immune function in detecting and clearing pulmonary pathogens; DC-SIGN is involved in facilitating dendritic cell interaction with naïve T cells to mount an anti-viral immune response. SP-D and DC-SIGN have been shown to interact with various viruses, including SARS-CoV-2, an enveloped RNA virus that causes COVID-19. A recombinant fragment of human SP-D (rfhSP-D) comprising of α-helical neck region, carbohydrate recognition domain, and eight N-terminal Gly-X-Y repeats has been shown to bind SARS-CoV-2 Spike protein and inhibit SARS-CoV-2 replication by preventing viral entry in Vero cells and HEK293T cells expressing ACE2. DC-SIGN has also been shown to act as a cell surface receptor for SARS-CoV-2 independent of ACE2. Since rfhSP-D is known to interact with SARS-CoV-2 Spike protein and DC-SIGN, this study was aimed at investigating the potential of rfhSP-D in modulating SARS-CoV-2 infection. Coincubation of rfhSP-D with Spike protein improved the Spike Protein: DC-SIGN interaction. Molecular dynamic studies revealed that rfhSP-D stabilised the interaction between DC-SIGN and Spike protein. Cell binding analysis with DC-SIGN expressing HEK 293T and THP- 1 cells and rfhSP-D treated SARS-CoV-2 Spike pseudotypes confirmed the increased binding. Furthermore, infection assays using the pseudotypes revealed their increased uptake by DC-SIGN expressing cells. The immunomodulatory effect of rfhSP-D on the DC-SIGN: Spike protein interaction on DC-SIGN expressing epithelial and macrophage-like cell lines was also assessed by measuring the mRNA expression of cytokines and chemokines. RT-qPCR analysis showed that rfhSP-D treatment downregulated the mRNA expression levels of pro-inflammatory cytokines and chemokines such as TNF-α, IFN-α, IL-1β, IL- 6, IL-8, and RANTES (as well as NF-κB) in DC-SIGN expressing cells challenged by Spike protein. Furthermore, rfhSP-D treatment was found to downregulate the mRNA levels of MHC class II in DC expressing THP-1 when compared to the untreated controls. We conclude that rfhSP-D helps stabilise the interaction between SARS- CoV-2 Spike protein and DC-SIGN and increases viral uptake by macrophages via DC-SIGN, suggesting an additional role for rfhSP-D in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Nazar Beirag
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Chandan Kumar
- Biomedical Informatics Centre, National Institute for Research in Reproductive and Child Health, ICMR, Mumbai, Maharashtra, India
| | - Taruna Madan
- Department of Innate Immunity, National Institute for Research in Reproductive and Child Health, ICMR, Mumbai, India
| | - Mohamed H. Shamji
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute and NIHR Biomedical Research Centre, Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom
| | - Roberta Bulla
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Daniel Mitchell
- WMS - Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Valarmathy Murugaiah
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Martin Mayora Neto
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent and Greenwich, United Kingdom
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent and Greenwich, United Kingdom
| | - Susan Idicula-Thomas
- Biomedical Informatics Centre, National Institute for Research in Reproductive and Child Health, ICMR, Mumbai, Maharashtra, India
| | - Praveen M. Varghese
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
- *Correspondence: Praveen M. Varghese, ; Uday Kishore,
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
- Department of Veterinary Medicine, U.A.E. University, Al Ain, United Arab Emirates
- *Correspondence: Praveen M. Varghese, ; Uday Kishore,
| |
Collapse
|
12
|
Fang S, Ju D, Lin Y, Chen W. The role of interleukin-22 in lung health and its therapeutic potential for COVID-19. Front Immunol 2022; 13:951107. [PMID: 35967401 PMCID: PMC9364265 DOI: 10.3389/fimmu.2022.951107] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Although numerous clinical trials have been implemented, an absolutely effective treatment against coronavirus disease 2019 (COVID-19) is still elusive. Interleukin-22 (IL-22) has attracted great interest over recent years, making it one of the best-studied cytokines of the interleukin-10 (IL-10) family. Unlike most interleukins, the major impact of IL-22 is exclusively on fibroblasts and epithelial cells due to the restricted expression of receptor. Numerous studies have suggested that IL-22 plays a crucial role in anti-viral infections through significantly ameliorating the immune cell-mediated inflammatory responses, and reducing tissue injury as well as further promoting epithelial repair and regeneration. Herein, we pay special attention to the role of IL-22 in the lungs. We summarize the latest progress in our understanding of IL-22 in lung health and disease and further discuss maneuvering this cytokine as potential immunotherapeutic strategy for the effective manage of COVID-19.
Collapse
Affiliation(s)
- Si Fang
- Multiscale Research Institute of Complex Systems & Jingan District Central Hospital of Shanghai, Fudan University, Shanghai, China
| | - Dianwen Ju
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, Fudan University School of Pharmacy, Shanghai, China
| | - Yong Lin
- Multiscale Research Institute of Complex Systems & Jingan District Central Hospital of Shanghai, Fudan University, Shanghai, China
| | - Wei Chen
- Multiscale Research Institute of Complex Systems & Jingan District Central Hospital of Shanghai, Fudan University, Shanghai, China
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, United States
| |
Collapse
|
13
|
McCreary MR, Schnell PM, Rhoda DA. Randomized double-blind placebo-controlled proof-of-concept trial of resveratrol for outpatient treatment of mild coronavirus disease (COVID-19). Sci Rep 2022; 12:10978. [PMID: 35768453 PMCID: PMC9243086 DOI: 10.1038/s41598-022-13920-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 05/30/2022] [Indexed: 12/13/2022] Open
Abstract
Resveratrol is a polyphenol that has been well studied and has demonstrated anti-viral and anti-inflammatory properties that might mitigate the effects of COVID-19. Outpatients (N = 105) were recruited from central Ohio in late 2020. Participants were randomly assigned to receive placebo or resveratrol. Both groups received a single dose of Vitamin D3 which was used as an adjunct. The primary outcome measure was hospitalization within 21 days of symptom onset; secondary measures were ER visits, incidence of pneumonia, and incidence of pulmonary embolism. Five patients chose not to participate after randomization. Twenty-one-day outcome was determined of all one hundred participants (mean [SD] age 55.6 [8.8] years; 61% female). There were no clinically significant adverse events attributed to resveratrol. Outpatients in this phase 2 study treated with resveratrol had a lower incidence compared to placebo of: hospitalization (2% vs. 6%, RR 0.33, 95% CI 0.04-3.10), COVID-19 related ER visits (8% vs. 14%, RR 0.57, 95% CI 0.18-1.83), and pneumonia (8% vs. 16%, RR 0.5, 95% CI 0.16-1.55). One patient (2%) in each group developed pulmonary embolism (RR 1.00, 95% CI: 0.06-15.55). This underpowered study was limited by small sample size and low incidence of primary adverse events consequently the results are statistically similar between treatment arms. A larger trial could determine efficacy.Trial Registrations: ClinicalTrials.gov NCT04400890 26/05/2020; FDA IND #150033 05/05/2020.
Collapse
Affiliation(s)
- Marvin R McCreary
- Department of Emergency Medicine, Mount Carmel Health Systems, Columbus, OH, 43213, USA.
| | - Patrick M Schnell
- Division of Biostatistics, College of Public Health, The Ohio State University, Columbus, OH, 43210, USA
| | - Dale A Rhoda
- Biostat Global Consulting, Worthington, OH, 43085, USA
| |
Collapse
|
14
|
Dogan S, Mart Komurcu SZ, Korkmaz MD, Kaya E, Yavas S, Dogan S, Senturk Ciftci H, Dasdemir S. Effect of Chemokine Gene Variants on Covid-19 Disease Severity. Immunol Invest 2022; 51:1965-1974. [PMID: 35763308 DOI: 10.1080/08820139.2022.2088383] [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: 11/05/2022]
Abstract
Patients immune phenotype/genotype data may be useful to understand the molecular mechanisms involved in SARS-CoV-2 infection and can contribute to the identify the different levels of disease severity. The roles of chemokines have been reported in the coronavirus-related diseases SARS and MERS and they may likewise play a critical role in the development of the symptoms of COVID-19 disease. We analyzed the association of the MCP-1-A2518 G, SDF-1-3'A, CCR5-delta32, CCR5-A55029 G, CXCR4-C138T and CCR2-V64I gene polymorphisms with COVID-19 severity to further unveil the immunological pathways leading to disease severity and death. Polymerase chain reaction(PCR)/Sanger sequencing analysis was performed for detection of the variations in 60 asymptomatic and 119 severe COVID-19 patients. In our study, we found that the frequencies of MCP-1 of GA genotype and G allele carriers were significantly higher in severe COVID-19 patients than the asymptomatic COVID-19 patients (p < .0001 and p: .005, respectively). However, no significant association was found for any of the other polymorphisms with the severity of COVID-19. Our findings suggest that there is a positive association between MCP-1-A2518 G gene variants with the severity of COVID-19. However, larger studies in different population which will focus on gene expression levels will help us to understand the capability of the mechanism role.
Collapse
Affiliation(s)
- Seydanur Dogan
- Istanbul Faculty of Medicine, Department of Medical Biology, Istanbul University, Istanbul, Turkey
| | | | - Merve Damla Korkmaz
- Department of Physical Medicine and Rehabilitation, Kanuni Sultan Suleyman Training and Research Hospital, Istanbul, Turkey
| | - Ebru Kaya
- Department of Anesthesiology and Reanimation, Kanuni Sultan Suleyman Training and Research Hospital, Istanbul, Turkey
| | - Sevim Yavas
- Department of Infectious Diseases, Kanuni Sultan Suleyman Training and Research Hospital, Istanbul, Turkey
| | - Serkan Dogan
- Department of Emergency Medicine, Kanuni Sultan Suleyman Training and Research Hospital, Istanbul, Turkey
| | - Hayriye Senturk Ciftci
- Istanbul Faculty of Medicine, Department of Medical Biology, Istanbul University, Istanbul, Turkey
| | - Selcuk Dasdemir
- Istanbul Faculty of Medicine, Department of Medical Biology, Istanbul University, Istanbul, Turkey
| |
Collapse
|
15
|
Li H, Wu M, Zhao X. Role of chemokine systems in cancer and inflammatory diseases. MedComm (Beijing) 2022; 3:e147. [PMID: 35702353 PMCID: PMC9175564 DOI: 10.1002/mco2.147] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 12/12/2022] Open
Abstract
Chemokines are a large family of small secreted proteins that have fundamental roles in organ development, normal physiology, and immune responses upon binding to their corresponding receptors. The primary functions of chemokines are to coordinate and recruit immune cells to and from tissues and to participate in regulating interactions between immune cells. In addition to the generally recognized antimicrobial immunity, the chemokine/chemokine receptor axis also exerts a tumorigenic function in many different cancer models and is involved in the formation of immunosuppressive and protective tumor microenvironment (TME), making them potential prognostic markers for various hematologic and solid tumors. In fact, apart from its vital role in tumors, almost all inflammatory diseases involve chemokines and their receptors in one way or another. Modulating the expression of chemokines and/or their corresponding receptors on tumor cells or immune cells provides the basis for the exploitation of new drugs for clinical evaluation in the treatment of related diseases. Here, we summarize recent advances of chemokine systems in protumor and antitumor immune responses and discuss the prevailing understanding of how the chemokine system operates in inflammatory diseases. In this review, we also emphatically highlight the complexity of the chemokine system and explore its potential to guide the treatment of cancer and inflammatory diseases.
Collapse
Affiliation(s)
- Hongyi Li
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of EducationWest China Second HospitalSichuan UniversityChengduChina
| | - Min Wu
- Department of Biomedical Sciences, School of Medicine and Health SciencesUniversity of North DakotaGrand ForksNorth DakotaUSA
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of EducationWest China Second HospitalSichuan UniversityChengduChina
| |
Collapse
|
16
|
Park SJ, Nam DE, Seong HC, Hahn YS. New Discovery of Myeloid-Derived Suppressor Cell's Tale on Viral Infection and COVID-19. Front Immunol 2022; 13:842535. [PMID: 35185933 PMCID: PMC8850309 DOI: 10.3389/fimmu.2022.842535] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/17/2022] [Indexed: 01/08/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are generated under biological stress such as cancer, inflammatory tissue damage, and viral infection. In recent years, with occurrence of global infectious diseases, new discovery on MDSCs functions has been significantly expanded during viral infection and COVID-19. For a successful viral infection, pathogens viruses develop immune evasion strategies to avoid immune recognition. Numerous viruses induce the differentiation and expansion of MDSCs in order to suppress host immune responses including natural killer cells, antigen presenting cells, and T-cells. Moreover, MDSCs play an important role in regulation of immunopathogenesis by balancing viral infection and tissue damage. In this review article, we describe the overview of immunomodulation and genetic regulation of MDSCs during viral infection in the animal model and human studies. In addition, we include up-to-date review of role of MDSCs in SARS-CoV-2 infection and COVID-19. Finally, we discuss potential therapeutics targeting MDSCs.
Collapse
Affiliation(s)
- Soo-Jeung Park
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Da-eun Nam
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Hae Chang Seong
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Young S. Hahn
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
17
|
Zepeda-Cervantes J, Martínez-Flores D, Ramírez-Jarquín JO, Tecalco-Cruz ÁC, Alavez-Pérez NS, Vaca L, Sarmiento-Silva RE. Implications of the Immune Polymorphisms of the Host and the Genetic Variability of SARS-CoV-2 in the Development of COVID-19. Viruses 2022; 14:94. [PMID: 35062298 PMCID: PMC8778858 DOI: 10.3390/v14010094] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/22/2021] [Accepted: 12/28/2021] [Indexed: 01/08/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is responsible for the current pandemic affecting almost all countries in the world. SARS-CoV-2 is the agent responsible for coronavirus disease 19 (COVID-19), which has claimed millions of lives around the world. In most patients, SARS-CoV-2 infection does not cause clinical signs. However, some infected people develop symptoms, which include loss of smell or taste, fever, dry cough, headache, severe pneumonia, as well as coagulation disorders. The aim of this work is to report genetic factors of SARS-CoV-2 and host-associated to severe COVID-19, placing special emphasis on the viral entry and molecules of the immune system involved with viral infection. Besides this, we analyze SARS-CoV-2 variants and their structural characteristics related to the binding to polymorphic angiotensin-converting enzyme type 2 (ACE2). Additionally, we also review other polymorphisms as well as some epigenetic factors involved in the immunopathogenesis of COVID-19. These factors and viral variability could explain the increment of infection rate and/or in the development of severe COVID-19.
Collapse
Affiliation(s)
- Jesús Zepeda-Cervantes
- Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Daniel Martínez-Flores
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Josué Orlando Ramírez-Jarquín
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Ángeles C. Tecalco-Cruz
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México (UACM), Mexico City 06720, Mexico;
| | - Noé Santiago Alavez-Pérez
- Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Mexico City 07340, Mexico;
| | - Luis Vaca
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Rosa Elena Sarmiento-Silva
- Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| |
Collapse
|
18
|
Xie P, Fang Y, Shen Z, Shao Y, Ma Q, Yang Z, Zhao J, Li H, Li R, Dong S, Wen W, Xia X. Broad antiviral and anti-inflammatory activity of Qingwenjiere mixture against SARS-CoV-2 and other human coronavirus infections. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 93:153808. [PMID: 34753027 PMCID: PMC8522682 DOI: 10.1016/j.phymed.2021.153808] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 10/07/2021] [Accepted: 10/17/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Qingwenjiere Mixture (QJM) is a traditional Chinese medicine (TCM) that has been shown to have remarkable clinical efficacy against COVID-19. However, little is known about the antiviral and anti-inflammatory activities of QJM against a wider range of human coronavirus (HCoV) strains. PURPOSE The study aims to investigate the antiviral and anti-inflammatory activities of QJM, as well as the underlying mechanisms against HCoV infections. METHODS The chemical compositions from QJM were analyzed by LC-MS. The inhibitory effect of QJM on infections of HCoV-OC43, HCoV-229E, HCoV-NL63, and SARS-CoV-2 was evaluated in HRT-18 cells, Huh7 cells, LLC-MK2 cells, and Vero-E6 cells, respectively, by using cytopathic effect (CPE) inhibition assay or RT-qPCR detection of viral n, s, or RdRp/Hel genes. The expression of pro-inflammatory cytokines induced by HCoV-OC43, HCoV-229E, and SARS-CoV-2, as well as the host ace2 gene was also determined by RT-qPCR assay. Furthermore, the expression of key molecules in the NF-κB/MAPKs signaling pathways was determined by western blot. RESULTS In alcohol-extraction groups of QJM and reference decoction pieces, 53 similar ion peaks were identified, the majority of which were phenylpropanoids, iridoids, and flavonoids. In addition, QJM reduced CPE caused by HCoVs and the expression of viral n genes or N protein. Pretreatment with QJM also exerted inhibitory effect on viral n gene expression. QJM also inhibited the expression of RdRp/Hel and s genes of SARS-CoV-2, as well as the host ace2 gene. Besides, QJM markedly reduced virus-induced mRNA expression of a panel of pro-inflammatory cytokines, such as IL-6, CXCL-8/IL-8, CXCL-10/IP-10, CCL-5/RANTES, TNF-α, IFN-α, CCL-2/MCP-1, CXCL-9/MIG, and IL1-α. We further showed that QJM inhibited the phosphorylation of NF-κB p65, and JNK, ERK 1/2, and p38 MAPKs in HCoV-OC43-infected HRT-18 cells. CONCLUSIONS QJM has broad antiviral and anti-inflammatory activity against both common and newly emerged HCoVs possibly by inhibiting the activation of key components in NF-κB/MAPKs signaling pathway. QJM also has a prevention effect against HCoV infections and inhibits the host receptor required for virus entry. These results indicate that QJM may have the therapeutic potential in the treatment of diseases caused by a broad range of HCoVs.
Collapse
Affiliation(s)
- Peifang Xie
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Yue Fang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Zhili Shen
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Yulan Shao
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Qinhai Ma
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Zifeng Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Hongmei Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Rongtao Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Shuwei Dong
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China.
| | - Weibo Wen
- Chinese medicine hospitals in Yunnan Province, Yunnan University of Chinese medicine, Kunming, Yunnan 650500, China.
| | - Xueshan Xia
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China.
| |
Collapse
|
19
|
Teixeira PC, Dorneles GP, Santana Filho PC, da Silva IM, Schipper LL, Postiga IAL, Neves CAM, Rodrigues Junior LC, Peres A, Souto JTD, Fonseca SG, Eller S, Oliveira TF, Rotta LN, Thompson CE, Romão PRT. Increased LPS levels coexist with systemic inflammation and result in monocyte activation in severe COVID-19 patients. Int Immunopharmacol 2021; 100:108125. [PMID: 34543980 PMCID: PMC8426217 DOI: 10.1016/j.intimp.2021.108125] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/28/2021] [Accepted: 08/31/2021] [Indexed: 12/23/2022]
Abstract
Mucosal barrier alterations may play a role in the pathogenesis of several diseases, including COVID-19. In this study we evaluate the association between bacterial translocation markers and systemic inflammation at the earliest time-point after hospitalization and at the last 72 h of hospitalization in survivors and non-survivors COVID-19 patients. Sixty-six SARS-CoV-2 RT-PCR positive patients and nine non-COVID-19 pneumonia controls were admitted in this study. Blood samples were collected at hospital admission (T1) (Controls and COVID-19 patients) and 0-72 h before hospital discharge (T2, alive or dead) to analyze systemic cytokines and chemokines, lipopolysaccharide (LPS) concentrations and soluble CD14 (sCD14) levels. THP-1 human monocytic cell line was incubated with plasma from survivors and non-survivors COVID-19 patients and their phenotype, activation status, TLR4, and chemokine receptors were analyzed by flow cytometry. COVID-19 patients presented higher IL-6, IFN-γ, TNF-α, TGF-β1, CCL2/MCP-1, CCL4/MIP-1β, and CCL5/RANTES levels than controls. Moreover, LPS and sCD14 were higher at hospital admission in SARS-CoV-2-infected patients. Non-survivors COVID-19 patients had increased LPS levels concomitant with higher IL-6, TNF-α, CCL2/MCP-1, and CCL5/RANTES levels at T2. Increased expression of CD16 and CCR5 were identified in THP-1 cells incubated with the plasma of survivor patients obtained at T2. The incubation of THP-1 with T2 plasma of non-survivors COVID-19 leads to higher TLR4, CCR2, CCR5, CCR7, and CD69 expression. In conclusion, the coexistence of increased microbial translocation and hyperinflammation in patients with severe COVID-19 may lead to higher monocyte activation, which may be associated with worsening outcomes, such as death.
Collapse
Affiliation(s)
- Paula C Teixeira
- Laboratory of Cellular and Molecular Immunology, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil; Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
| | - Gilson P Dorneles
- Laboratory of Cellular and Molecular Immunology, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil; Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil.
| | - Paulo C Santana Filho
- Laboratory of Cellular and Molecular Immunology, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
| | - Igor M da Silva
- Laboratory of Cellular and Molecular Immunology, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
| | - Lucas L Schipper
- Laboratory of Cellular and Molecular Immunology, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil; Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
| | - Isabelle A L Postiga
- Laboratory of Cellular and Molecular Immunology, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
| | - Carla Andretta Moreira Neves
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
| | | | - Alessandra Peres
- Laboratory of Cellular and Molecular Immunology, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil; Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil; Graduate Program in Rehabilitation Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
| | - Janeusa Trindade de Souto
- Department of Microbiology and Parasitology, Federal University of Rio Grande do Norte, Natal, Brazil
| | | | - Sarah Eller
- Pharmacosciences Department, Federal University of Health Sciences of Porto Alegre, Brazil
| | - Tiago F Oliveira
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil; Pharmacosciences Department, Federal University of Health Sciences of Porto Alegre, Brazil
| | - Liane N Rotta
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
| | - Claudia Elizabeth Thompson
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil; Pharmacosciences Department, Federal University of Health Sciences of Porto Alegre, Brazil
| | - Pedro R T Romão
- Laboratory of Cellular and Molecular Immunology, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil; Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil; Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil.
| |
Collapse
|
20
|
Christie MJ, Irving AT, Forster SC, Marsland BJ, Hansbro PM, Hertzog PJ, Nold-Petry CA, Nold MF. Of bats and men: Immunomodulatory treatment options for COVID-19 guided by the immunopathology of SARS-CoV-2 infection. Sci Immunol 2021; 6:eabd0205. [PMID: 34533977 DOI: 10.1126/sciimmunol.abd0205] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Michael J Christie
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia
| | - Aaron T Irving
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining 314400, China
| | - Samuel C Forster
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, Victoria, Australia.,Department of Molecular and Translational Sciences, Monash University, Melbourne, Victoria, Australia
| | - Benjamin J Marsland
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute, Sydney, NSW 2050, Australia.,Centre for Inflammation, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Paul J Hertzog
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, Victoria, Australia.,Department of Molecular and Translational Sciences, Monash University, Melbourne, Victoria, Australia
| | - Claudia A Nold-Petry
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia
| | - Marcel F Nold
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia.,Monash Newborn, Monash Children's Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
21
|
Methylation of Host Genes Associated with Coronavirus Infection from Birth to 26 Years. Genes (Basel) 2021; 12:genes12081198. [PMID: 34440372 PMCID: PMC8392033 DOI: 10.3390/genes12081198] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 07/26/2021] [Accepted: 07/29/2021] [Indexed: 12/13/2022] Open
Abstract
DNA methylation (DNAm) patterns over time at 1146 CpGs on coronavirus-related genes were assessed to understand whether the varying differences in susceptibility, symptoms, and the outcomes of the SARS-CoV-2 infection in children and young adults could be explained through epigenetic alterations in a host cell’s transcriptional apparatus to coronaviruses. DNAm data from the Isle of Wight birth cohort (IOWBC) at birth, 10, 18, and 26 years of age were included. Linear mixed models with repeated measurements stratified by sex were used to examine temporal patterns, and cluster analysis was performed to identify CpGs following similar patterns. CpGs on autosomes and sex chromosomes were analyzed separately. The association of identified CpGs and expression of their genes were evaluated. Pathway enrichment analyses of the genes was conducted at FDR = 0.05. DNAm at 635 of the 1146 CpGs on autosomes showed statistically significant time effects (FDR = 0.05). The 635 CpGs were classified into five clusters with each representing a unique temporal pattern of DNAm. Of the 29 CpGs on sex chromosomes, DNAm at seven CpGs in males and eight CpGs in females showed time effects (FDR = 0.05). Sex-specific and non-specific associations of DNAm with gene expression were found at 24 and 93 CpGs, respectively. Genes which mapped the 643 CpGs represent 460 biological processes. We suggest that the observed variability in DNAm with advancing age may partially explain differing susceptibility, disease severity, and mortality of coronavirus infections among different age groups.
Collapse
|
22
|
Rowlands M, Segal F, Hartl D. Myeloid-Derived Suppressor Cells as a Potential Biomarker and Therapeutic Target in COVID-19. Front Immunol 2021; 12:697405. [PMID: 34220859 PMCID: PMC8250151 DOI: 10.3389/fimmu.2021.697405] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/07/2021] [Indexed: 12/11/2022] Open
Abstract
Clinical presentations of COVID-19 are highly variable, yet the precise mechanisms that govern the pathophysiology of different disease courses remain poorly defined. Across the spectrum of disease severity, COVID-19 impairs both innate and adaptive host immune responses by activating innate immune cell recruitment, while resulting in low lymphocyte counts. Recently, several reports have shown that patients with severe COVID-19 exhibit a dysregulated myeloid cell compartment, with increased myeloid-derived suppressor cells (MDSCs) correlating with disease severity. MDSCs, in turn, promote virus survival by suppressing T-cell responses and driving a highly pro-inflammatory state through the secretion of various mediators of immune activation. Here, we summarize the evidence on MDSCs and myeloid cell dysregulation in COVID-19 infection and discuss the potential of MDSCs as biomarkers and therapeutic targets in COVID-19 pneumonia and associated disease.
Collapse
Affiliation(s)
- Marianna Rowlands
- Novartis Institutes for BioMedical Research (NIBR) Translational Medicine, Cambridge, MA, United States
| | - Florencia Segal
- Novartis Institutes for BioMedical Research (NIBR) Translational Medicine, Cambridge, MA, United States
| | - Dominik Hartl
- Novartis Institutes for BioMedical Research (NIBR), Translational Medicine, Basel, Switzerland.,Department of Pediatrics I, University of Tübingen, Tübingen, Germany
| |
Collapse
|
23
|
Järhult JD, Hultström M, Bergqvist A, Frithiof R, Lipcsey M. The impact of viremia on organ failure, biomarkers and mortality in a Swedish cohort of critically ill COVID-19 patients. Sci Rep 2021; 11:7163. [PMID: 33785784 PMCID: PMC8010103 DOI: 10.1038/s41598-021-86500-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 03/16/2021] [Indexed: 12/15/2022] Open
Abstract
The spread of virus via the blood stream has been suggested to contribute to extra-pulmonary organ failure in Coronavirus disease 2019 (COVID-19). We assessed SARS-CoV-2 RNAemia (RNAemia) and the association between RNAemia and inflammation, organ failure and mortality in critically ill COVID-19 patients. We included all patients with PCR verified COVID-19 and consent admitted to ICU. SARS-CoV-2 RNA copies above 1000/ml measured by PCR in plasma was defined as RNAemia and used as surrogate for viremia. In this cohort of 92 patients 59 (64%) were invasively ventilated. RNAemia was found in 31 patients (34%). Hypertension and corticosteroid treatment was more common in patients with RNAemia. Extra-pulmonary organ failure biomarkers and the extent of organ failure were similar in patients with and without RNAemia, but the former group had more renal replacement therapy and higher mortality (26 vs 16%; 35 vs 16%, respectively, p = 0.04). RNAemia was not an independent predictor of death at 30 days after adjustment for age. SARS-CoV2 RNA copies in plasma is a common finding in ICU patients with COVID-19. Although viremia was not associated with extra pulmonary organ failure it was more common in patients who did not survive to 30 days after ICU admission.Trial registration: ClinicalTrials NCT04316884.
Collapse
Affiliation(s)
- Josef D Järhult
- Department of Medical Sciences, Zoonosis Science Center, Uppsala University, Uppsala, Sweden
| | - Michael Hultström
- Department of Surgical Sciences, Anesthesia and Intensive Care Medicine, Uppsala University, Uppsala, Sweden
- Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Anders Bergqvist
- Clinical Microbiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Clinical Microbiology and Hospital Infection Control, Uppsala University Hospital, Uppsala, Sweden
| | - Robert Frithiof
- Department of Surgical Sciences, Anesthesia and Intensive Care Medicine, Uppsala University, Uppsala, Sweden
| | - Miklos Lipcsey
- Department of Surgical Sciences, Anesthesia and Intensive Care Medicine, Uppsala University, Uppsala, Sweden.
- Hedenstierna Laboratory, CIRRUS, Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
24
|
Daamen AR, Bachali P, Owen KA, Kingsmore KM, Hubbard EL, Labonte AC, Robl R, Shrotri S, Grammer AC, Lipsky PE. Comprehensive transcriptomic analysis of COVID-19 blood, lung, and airway. Sci Rep 2021; 11:7052. [PMID: 33782412 PMCID: PMC8007747 DOI: 10.1038/s41598-021-86002-x] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 03/03/2021] [Indexed: 02/01/2023] Open
Abstract
SARS-CoV2 is a previously uncharacterized coronavirus and causative agent of the COVID-19 pandemic. The host response to SARS-CoV2 has not yet been fully delineated, hampering a precise approach to therapy. To address this, we carried out a comprehensive analysis of gene expression data from the blood, lung, and airway of COVID-19 patients. Our results indicate that COVID-19 pathogenesis is driven by populations of myeloid-lineage cells with highly inflammatory but distinct transcriptional signatures in each compartment. The relative absence of cytotoxic cells in the lung suggests a model in which delayed clearance of the virus may permit exaggerated myeloid cell activation that contributes to disease pathogenesis by the production of inflammatory mediators. The gene expression profiles also identify potential therapeutic targets that could be modified with available drugs. The data suggest that transcriptomic profiling can provide an understanding of the pathogenesis of COVID-19 in individual patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Robert Robl
- AMPEL BioSolutions LLC, Charlottesville, VA, 22902, USA
| | - Sneha Shrotri
- AMPEL BioSolutions LLC, Charlottesville, VA, 22902, USA
| | | | - Peter E Lipsky
- AMPEL BioSolutions LLC, Charlottesville, VA, 22902, USA.
| |
Collapse
|
25
|
Sur S, Khatun M, Steele R, Isbell TS, Ray R, Ray RB. Exosomes from COVID-19 Patients Carry Tenascin-C and Fibrinogen-β in Triggering Inflammatory Signals in Cells of Distant Organ. Int J Mol Sci 2021; 22:ijms22063184. [PMID: 33804769 PMCID: PMC8003878 DOI: 10.3390/ijms22063184] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/17/2021] [Accepted: 03/17/2021] [Indexed: 02/06/2023] Open
Abstract
SARS-CoV-2 infection can cause cytokine storm and may overshoot immunity in humans; however, it remains to be determined whether virus-induced soluble mediators from infected cells are carried by exosomes as vehicles to distant organs and cause tissue damage in COVID-19 patients. We took an unbiased proteomic approach for analyses of exosomes isolated from plasma of healthy volunteers and COVID-19 patients. Our results revealed that tenascin-C (TNC) and fibrinogen-β (FGB) are highly abundant in exosomes from COVID-19 patients’ plasma compared with that of healthy normal controls. Since TNC and FGB stimulate pro-inflammatory cytokines via the Nuclear factor-κB (NF-κB) pathway, we examined the status of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and C–C motif chemokine ligand 5 (CCL5) expression upon exposure of hepatocytes to exosomes from COVID-19 patients and observed significant increase compared with that from healthy subjects. Together, our results demonstrate that TNC and FGB are transported through plasma exosomes and potentially trigger pro-inflammatory cytokine signaling in cells of distant organ.
Collapse
Affiliation(s)
- Subhayan Sur
- Department of Pathology, Saint Louis University, St. Louis, MO 63104, USA; (S.S.); (M.K.); (R.S.); (T.S.I.)
| | - Mousumi Khatun
- Department of Pathology, Saint Louis University, St. Louis, MO 63104, USA; (S.S.); (M.K.); (R.S.); (T.S.I.)
| | - Robert Steele
- Department of Pathology, Saint Louis University, St. Louis, MO 63104, USA; (S.S.); (M.K.); (R.S.); (T.S.I.)
| | - T. Scott Isbell
- Department of Pathology, Saint Louis University, St. Louis, MO 63104, USA; (S.S.); (M.K.); (R.S.); (T.S.I.)
| | - Ranjit Ray
- Department of Internal Medicine, Saint Louis University, St. Louis, MO 63104, USA;
| | - Ratna B. Ray
- Department of Pathology, Saint Louis University, St. Louis, MO 63104, USA; (S.S.); (M.K.); (R.S.); (T.S.I.)
- Correspondence:
| |
Collapse
|
26
|
Khalil BA, Elemam NM, Maghazachi AA. Chemokines and chemokine receptors during COVID-19 infection. Comput Struct Biotechnol J 2021; 19:976-988. [PMID: 33558827 PMCID: PMC7859556 DOI: 10.1016/j.csbj.2021.01.034] [Citation(s) in RCA: 135] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 12/17/2022] Open
Abstract
Chemokines are crucial inflammatory mediators needed during an immune response to clear pathogens. However, their excessive release is the main cause of hyperinflammation. In the recent COVID-19 outbreak, chemokines may be the direct cause of acute respiratory disease syndrome, a major complication leading to death in about 40% of severe cases. Several clinical investigations revealed that chemokines are directly involved in the different stages of SARS-CoV-2 infection. Here, we review the role of chemokines and their receptors in COVID-19 pathogenesis to better understand the disease immunopathology which may aid in developing possible therapeutic targets for the infection.
Collapse
Key Words
- AECs, airway epithelial cells
- AP-1, Activator Protein 1
- ARDS
- ARDS, acute respiratory disease syndrome
- BALF, bronchial alveolar lavage fluid
- CAP, community acquired pneumonia
- COVID-19
- CRS, cytokine releasing syndrome
- Chemokine Receptors
- Chemokines
- DCs, dendritic cells
- ECM, extracellular matrix
- GAGs, glycosaminoglycans
- HIV, human immunodeficiency virus
- HRSV, human respiratory syncytial virus
- IFN, interferon
- IMM, inflammatory monocytes and macrophages
- IP-10, IFN-γ-inducible protein 10
- IRF, interferon regulatory factor
- Immunity
- MERS-CoV, Middle East respiratory syndrome coronavirus
- NETs, neutrophil extracellular traps
- NF-κB, Nuclear Factor kappa-light-chain-enhancer of activated B cells
- NK cells, natural killer cells
- PBMCs, peripheral blood mononuclear cells
- PRR, pattern recognition receptors
- RSV, rous sarcoma virus
- SARS-CoV, severe acute respiratory syndrome coronavirus
- SARS-CoV-2
- TLR, toll like receptor
- TRIF, TIR-domain-containing adapter-inducing interferon-β
Collapse
Affiliation(s)
- Bariaa A. Khalil
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Immuno-Oncology Group, Sharjah Institute for Medical Research (SIMR), Sharjah, United Arab Emirates
| | - Noha Mousaad Elemam
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Immuno-Oncology Group, Sharjah Institute for Medical Research (SIMR), Sharjah, United Arab Emirates
| | - Azzam A. Maghazachi
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Immuno-Oncology Group, Sharjah Institute for Medical Research (SIMR), Sharjah, United Arab Emirates
| |
Collapse
|
27
|
Forbester JL, Humphreys IR. Genetic influences on viral-induced cytokine responses in the lung. Mucosal Immunol 2021; 14:14-25. [PMID: 33184476 PMCID: PMC7658619 DOI: 10.1038/s41385-020-00355-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/14/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023]
Abstract
Infection with respiratory viruses such as influenza, respiratory syncytial virus and coronavirus provides a difficult immunological challenge for the host, where a balance must be established between controlling viral replication and limiting damage to the delicate lung structure. Although the genetic architecture of host responses to respiratory viral infections is not yet understood, it is clear there is underlying heritability that influences pathogenesis. Immune control of virus replication is essential in respiratory infections, but overt activation can enhance inflammation and disease severity. Cytokines initiate antiviral immune responses but are implicated in viral pathogenesis. Here, we discuss how host genetic variation may influence cytokine responses to respiratory viral infections and, based on our current understanding of the role that cytokines play in viral pathogenesis, how this may influence disease severity. We also discuss how induced pluripotent stem cells may be utilised to probe the mechanistic implications of allelic variation in genes in virus-induced inflammatory responses. Ultimately, this could help to design better immune modulators, stratify high risk patients and tailor anti-inflammatory treatments, potentially expanding the ability to treat respiratory virus outbreaks in the future.
Collapse
Affiliation(s)
- Jessica L Forbester
- Division of Infection and Immunity/Systems Immunity University Research Institute, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, CF14 4XN, UK.
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headley Way, Headington, Oxford, OX3 9DS, UK.
| | - Ian R Humphreys
- Division of Infection and Immunity/Systems Immunity University Research Institute, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, CF14 4XN, UK
| |
Collapse
|
28
|
Alon R, Sportiello M, Kozlovski S, Kumar A, Reilly EC, Zarbock A, Garbi N, Topham DJ. Leukocyte trafficking to the lungs and beyond: lessons from influenza for COVID-19. Nat Rev Immunol 2021; 21:49-64. [PMID: 33214719 PMCID: PMC7675406 DOI: 10.1038/s41577-020-00470-2] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2020] [Indexed: 01/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of coronavirus disease 2019 (COVID-19). Understanding of the fundamental processes underlying the versatile clinical manifestations of COVID-19 is incomplete without comprehension of how different immune cells are recruited to various compartments of virus-infected lungs, and how this recruitment differs among individuals with different levels of disease severity. As in other respiratory infections, leukocyte recruitment to the respiratory system in people with COVID-19 is orchestrated by specific leukocyte trafficking molecules, and when uncontrolled and excessive it results in various pathological complications, both in the lungs and in other organs. In the absence of experimental data from physiologically relevant animal models, our knowledge of the trafficking signals displayed by distinct vascular beds and epithelial cell layers in response to infection by SARS-CoV-2 is still incomplete. However, SARS-CoV-2 and influenza virus elicit partially conserved inflammatory responses in the different respiratory epithelial cells encountered early in infection and may trigger partially overlapping combinations of trafficking signals in nearby blood vessels. Here, we review the molecular signals orchestrating leukocyte trafficking to airway and lung compartments during primary pneumotropic influenza virus infections and discuss potential similarities to distinct courses of primary SARS-CoV-2 infections. We also discuss how an imbalance in vascular activation by leukocytes outside the airways and lungs may contribute to extrapulmonary inflammatory complications in subsets of patients with COVID-19. These multiple molecular pathways are potential targets for therapeutic interventions in patients with severe COVID-19.
Collapse
Affiliation(s)
- Ronen Alon
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel.
| | - Mike Sportiello
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Stav Kozlovski
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Ashwin Kumar
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Emma C Reilly
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Alexander Zarbock
- Department of Cellular Immunology, Institute of Experimental Immunology Medical Faculty, University of Bonn, Bonn, Germany
| | - Natalio Garbi
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - David J Topham
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
29
|
Hutchings CJ. Mini-review: antibody therapeutics targeting G protein-coupled receptors and ion channels. Antib Ther 2020; 3:257-264. [PMID: 33912796 PMCID: PMC7799348 DOI: 10.1093/abt/tbaa023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/09/2020] [Accepted: 11/14/2020] [Indexed: 12/13/2022] Open
Abstract
Antibodies are now well established as therapeutics with many additional advantages over small molecules and peptides relative to their selectivity, bioavailability, half-life and effector function. Major classes of membrane-associated protein targets include G protein-coupled receptors (GPCRs) and ion channels that are linked to a wide range of disease indications across all therapeutic areas. This mini-review summarizes the antibody target landscape for both GPCRs and ion channels as well as current progress in the respective research and development pipelines with some example case studies highlighted from clinical studies, including those being evaluated for the treatment of symptoms in COVID-19 infection.
Collapse
|
30
|
Ahmad T, Chaudhuri R, Joshi MC, Almatroudi A, Rahmani AH, Ali SM. COVID-19: The Emerging Immunopathological Determinants for Recovery or Death. Front Microbiol 2020; 11:588409. [PMID: 33335518 PMCID: PMC7736111 DOI: 10.3389/fmicb.2020.588409] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/19/2020] [Indexed: 01/08/2023] Open
Abstract
Hyperactivation of the host immune system during infection by SARS-CoV-2 is the leading cause of death in COVID-19 patients. It is also evident that patients who develop mild/moderate symptoms and successfully recover display functional and well-regulated immune response. Whereas a delayed initial interferon response is associated with severe disease outcome and can be the tipping point towards immunopathological deterioration, often preceding death in COVID-19 patients. Further, adaptive immune response during COVID-19 is heterogeneous and poorly understood. At the same time, some studies suggest activated T and B cell response in severe and critically ill patients and the presence of SARS-CoV2-specific antibodies. Thus, understanding this problem and the underlying molecular pathways implicated in host immune function/dysfunction is imperative to devise effective therapeutic interventions. In this comprehensive review, we discuss the emerging immunopathological determinants and the mechanism of virus evasion by the host cell immune system. Using the knowledge gained from previous respiratory viruses and the emerging clinical and molecular findings on SARS-CoV-2, we have tried to provide a holistic understanding of the host innate and adaptive immune response that may determine disease outcome. Considering the critical role of the adaptive immune system during the viral clearance, we have presented the molecular insights of the plausible mechanisms involved in impaired T cell function/dysfunction during various stages of COVID-19.
Collapse
Affiliation(s)
- Tanveer Ahmad
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Rituparna Chaudhuri
- Department of Molecular and Cellular Neuroscience, Neurovirology Section, National Brain Research Centre (NBRC), Haryana, India
| | - Mohan C. Joshi
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Science, Qassim University, Buraydah, Saudi Arabia
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Science, Qassim University, Buraydah, Saudi Arabia
| | - Syed Mansoor Ali
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
31
|
Tosif S, Neeland MR, Sutton P, Licciardi PV, Sarkar S, Selva KJ, Do LAH, Donato C, Quan Toh Z, Higgins R, Van de Sandt C, Lemke MM, Lee CY, Shoffner SK, Flanagan KL, Arnold KB, Mordant FL, Mulholland K, Bines J, Dohle K, Pellicci DG, Curtis N, McNab S, Steer A, Saffery R, Subbarao K, Chung AW, Kedzierska K, Burgner DP, Crawford NW. Immune responses to SARS-CoV-2 in three children of parents with symptomatic COVID-19. Nat Commun 2020; 11:5703. [PMID: 33177504 PMCID: PMC7658256 DOI: 10.1038/s41467-020-19545-8] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/20/2020] [Indexed: 12/27/2022] Open
Abstract
Compared to adults, children with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have predominantly mild or asymptomatic infections, but the underlying immunological differences remain unclear. Here, we describe clinical features, virology, longitudinal cellular, and cytokine immune profile, SARS-CoV-2-specific serology and salivary antibody responses in a family of two parents with PCR-confirmed symptomatic SARS-CoV-2 infection and their three children, who tested repeatedly SARS-CoV-2 PCR negative. Cellular immune profiles and cytokine responses of all children are similar to their parents at all timepoints. All family members have salivary anti-SARS-CoV-2 antibodies detected, predominantly IgA, that coincide with symptom resolution in 3 of 4 symptomatic members. Plasma from both parents and one child have IgG antibody against the S1 protein and virus-neutralizing activity detected. Using a systems serology approach, we demonstrate higher levels of SARS-CoV-2-specific antibody features of these family members compared to healthy controls. These data indicate that children can mount an immune response to SARS-CoV-2 without virological confirmation of infection, raising the possibility that immunity in children can prevent the establishment of SARS-CoV-2 infection. Relying on routine virological and serological testing may not identify exposed children, with implications for epidemiological and clinical studies across the life-span.
Collapse
Affiliation(s)
- Shidan Tosif
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia.
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia.
- Department of General Medicine, The Royal Children's Hospital, Melbourne, Victoria, Australia.
| | - Melanie R Neeland
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Philip Sutton
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Paul V Licciardi
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Sohinee Sarkar
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Kevin J Selva
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Lien Anh Ha Do
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Celeste Donato
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Zheng Quan Toh
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Rachel Higgins
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Carolien Van de Sandt
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Melissa M Lemke
- Department of Biomedical Engineering, University of Michigan, MI, USA
| | - Christina Y Lee
- Department of Biomedical Engineering, University of Michigan, MI, USA
| | | | - Katie L Flanagan
- Department of Infectious Diseases, Launceston General Hospital, Launceston, Tasmania, Australia
- School of Health Sciences and School of Medicine, University of Tasmania, Launceston, Tasmania, Australia
- Department of Immunology and Pathology, Monash University, Commercial Road, Melbourne, Victoria, Australia
- School of Health and Biomedical Science, RMIT University, Melbourne, Victoria, Australia
| | - Kelly B Arnold
- Department of Biomedical Engineering, University of Michigan, MI, USA
| | - Francesca L Mordant
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Kim Mulholland
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
- Infectious Diseases Unit, Department of General Medicine, The Royal Children's Hospital, Melbourne, Australia
| | - Julie Bines
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
- Department of Gastroenterology, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Kate Dohle
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Daniel G Pellicci
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
- Infectious Diseases Unit, Department of General Medicine, The Royal Children's Hospital, Melbourne, Australia
| | - Sarah McNab
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
- Department of General Medicine, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Andrew Steer
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
- Infectious Diseases Unit, Department of General Medicine, The Royal Children's Hospital, Melbourne, Australia
| | - Richard Saffery
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Kanta Subbarao
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Melbourne, Australia
| | - Amy W Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - David P Burgner
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
- Infectious Diseases Unit, Department of General Medicine, The Royal Children's Hospital, Melbourne, Australia
| | - Nigel W Crawford
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
- Department of General Medicine, The Royal Children's Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
32
|
Hussen J, Kandeel M, Hemida MG, Al-Mubarak AIA. Antibody-Based Immunotherapeutic Strategies for COVID-19. Pathogens 2020; 9:E917. [PMID: 33167401 PMCID: PMC7694378 DOI: 10.3390/pathogens9110917] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/30/2020] [Accepted: 11/03/2020] [Indexed: 02/06/2023] Open
Abstract
Global efforts to contain the coronavirus disease-2019 (COVID-19) include the development of novel preventive vaccines and effective therapeutics. Passive antibody therapies using convalescent plasma, SARS-CoV-2 (Severe-Acute-Respiratory-Syndrome-Corona-Virus-2)-specific neutralizing antibodies (NAbs), and the development of monoclonal antibodies (MAbs) are among the most promising strategies for prophylaxis and treatment of SARS-CoV-2 infections. In addition, several immunomodulatory antibodies acting via several mechanisms to boost the host immune defense against SARS-CoV-2 infection as well as to avoid the harmful overreaction of the immune system are currently under clinical trial. Our main objective is to present the current most up-to-date progress in some clinical trials registered at ClinicalTrials.gov. We highlight the pros and pitfalls of several SARS-CoV-2 antibody-based immunotherapeutics.
Collapse
Affiliation(s)
- Jamal Hussen
- Department of Microbiology, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (J.H.); (M.G.H.)
| | - Mahmoud Kandeel
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al-Hofuf, Al-Ahsa 31982, Saudi Arabia;
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelshikh University, Kafrelshikh 33516, Egypt
| | - Maged Gomaa Hemida
- Department of Microbiology, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (J.H.); (M.G.H.)
- Department of Virology, Faculty of Veterinary Medicine, Kafrelshikh University, Kafrelshikh 33516, Egypt
| | - Abdullah I. A. Al-Mubarak
- Department of Microbiology, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (J.H.); (M.G.H.)
| |
Collapse
|
33
|
Alnefaie A, Albogami S. Current approaches used in treating COVID-19 from a molecular mechanisms and immune response perspective. Saudi Pharm J 2020; 28:1333-1352. [PMID: 32905015 PMCID: PMC7462599 DOI: 10.1016/j.jsps.2020.08.024] [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: 06/23/2020] [Accepted: 08/27/2020] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), which is caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), was declared by the World Health Organization (WHO) as a global pandemic on March 11, 2020. SARS-CoV-2 targets the respiratory system, resulting in symptoms such as fever, headache, dry cough, dyspnea, and dizziness. These symptoms vary from person to person, ranging from mild to hypoxia with acute respiratory distress syndrome (ARDS) and sometimes death. Although not confirmed, phylogenetic analysis suggests that SARS-CoV-2 may have originated from bats; the intermediary facilitating its transfer from bats to humans is unknown. Owing to the rapid spread of infection and high number of deaths caused by SARS-CoV-2, most countries have enacted strict curfews and the practice of social distancing while awaiting the availability of effective U.S. Food and Drug Administration (FDA)-approved medications and/or vaccines. This review offers an overview of the various types of coronaviruses (CoVs), their targeted hosts and cellular receptors, a timeline of their emergence, and the roles of key elements of the immune system in fighting pathogen attacks, while focusing on SARS-CoV-2 and its genomic structure and pathogenesis. Furthermore, we review drugs targeting COVID-19 that are under investigation and in clinical trials, in addition to progress using mesenchymal stem cells to treat COVID-19. We conclude by reviewing the latest updates on COVID-19 vaccine development. Understanding the molecular mechanisms of how SARS-CoV-2 interacts with host cells and stimulates the immune response is extremely important, especially as scientists look for new strategies to guide their development of specific COVID-19 therapies and vaccines.
Collapse
Key Words
- ACE2, angiotensin-converting enzyme 2
- AHFS, American Hospital Formula Service
- ANGII, angiotensin II
- APCs, antigen presenting cells
- ARDS, acute respiratory distress syndrome
- COVID-19, coronavirus disease
- CoVs, coronaviruses
- Coronavirus
- GVHD, graft versus host disease
- HCoVs, human coronoaviruses
- IBV, infectious bronchitis coronavirus
- IFN-γ, interferon-gamma
- ILCs, innate lymphoid cells
- Investigational medications
- MERS-CoV, Middle East respiratory syndrome
- NKs, natural killer cells
- ORFs, open reading frames
- PAMPs, pathogen-associated molecular patterns
- Pandemic
- Pathophysiology
- RdRp, RNA-dependent RNA polymerase
- SARS-CoV-2
- SARS-CoV-2, severe acute respiratory syndrome coronavirus 2
- SLE, systemic lupus erythematosus
- TMPRSS2, transmembrane serine protease 2
- Viral immune response
- WHO, World Health Organization
- nsps, nonstructural proteins
Collapse
Affiliation(s)
- Alaa Alnefaie
- Department of Biotechnology, Faculty of Science, Taif University, Taif, Saudi Arabia
| | - Sarah Albogami
- Department of Biotechnology, Faculty of Science, Taif University, Taif, Saudi Arabia
| |
Collapse
|
34
|
Saha RP, Sharma AR, Singh MK, Samanta S, Bhakta S, Mandal S, Bhattacharya M, Lee SS, Chakraborty C. Repurposing Drugs, Ongoing Vaccine, and New Therapeutic Development Initiatives Against COVID-19. Front Pharmacol 2020; 11:1258. [PMID: 32973505 PMCID: PMC7466451 DOI: 10.3389/fphar.2020.01258] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 07/30/2020] [Indexed: 12/15/2022] Open
Abstract
As the COVID-19 is still growing throughout the globe, a thorough investigation into the specific immunopathology of SARS-CoV-2, its interaction with the host immune system and pathogen evasion mechanism may provide a clear picture of how the pathogen can breach the host immune defenses in elderly patients and patients with comorbid conditions. Such studies will also reveal the underlying mechanism of how children and young patients can withstand the disease better. The study of the immune defense mechanisms and the prolonged immune memory from patients population with convalescent plasma may help in designing a suitable vaccine candidate not only for the current outbreak but also for similar outbreaks in the future. The vital drug candidates, which are being tested as potential vaccines or therapeutics against COVID-19, include live attenuated vaccine, inactivated or killed vaccine, subunit vaccine, antibodies, interferon treatment, repurposing existing drugs, and nucleic acid-based vaccines. Several organizations around the world have fast-tracked the development of a COVID-19 vaccine, and some drugs already went to phase III of clinical trials. Hence, here, we have tried to take a quick glimpse of the development stages of vaccines or therapeutic approaches to treat this deadly disease.
Collapse
Affiliation(s)
- Rudra P. Saha
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata, India
| | - Ashish Ranjan Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, South Korea
| | - Manoj K. Singh
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata, India
| | - Saikat Samanta
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata, India
| | - Swarnav Bhakta
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata, India
| | - Snehasish Mandal
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata, India
| | - Manojit Bhattacharya
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, South Korea
| | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, South Korea
| | - Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science & Biotechnology, Adamas University, Kolkata, India
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, South Korea
| |
Collapse
|
35
|
Malik S, Gupta A, Zhong X, Rasmussen TP, Manautou JE, Bahal R. Emerging Therapeutic Modalities against COVID-19. Pharmaceuticals (Basel) 2020; 13:E188. [PMID: 32784499 PMCID: PMC7465781 DOI: 10.3390/ph13080188] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 07/30/2020] [Accepted: 08/04/2020] [Indexed: 02/06/2023] Open
Abstract
The novel SARS-CoV-2 virus has quickly spread worldwide, bringing the whole world as well as the economy to a standstill. As the world is struggling to minimize the transmission of this devastating disease, several strategies are being actively deployed to develop therapeutic interventions. Pharmaceutical companies and academic researchers are relentlessly working to investigate experimental, repurposed or FDA-approved drugs on a compassionate basis and novel biologics for SARS-CoV-2 prophylaxis and treatment. Presently, a tremendous surge of COVID-19 clinical trials are advancing through different stages. Among currently registered clinical efforts, ~86% are centered on testing small molecules or antibodies either alone or in combination with immunomodulators. The rest ~14% of clinical efforts are aimed at evaluating vaccines and convalescent plasma-based therapies to mitigate the disease's symptoms. This review provides a comprehensive overview of current therapeutic modalities being evaluated against SARS-CoV-2 virus in clinical trials.
Collapse
Affiliation(s)
- Shipra Malik
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA; (S.M.); (X.Z.); (T.P.R.); (J.E.M.)
| | - Anisha Gupta
- Department of Chemistry, Wesleyan University, Middletown, CT 06459, USA;
| | - Xiaobo Zhong
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA; (S.M.); (X.Z.); (T.P.R.); (J.E.M.)
| | - Theodore P. Rasmussen
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA; (S.M.); (X.Z.); (T.P.R.); (J.E.M.)
| | - Jose E. Manautou
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA; (S.M.); (X.Z.); (T.P.R.); (J.E.M.)
| | - Raman Bahal
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA; (S.M.); (X.Z.); (T.P.R.); (J.E.M.)
| |
Collapse
|
36
|
Valencia I, Peiró C, Lorenzo Ó, Sánchez-Ferrer CF, Eckel J, Romacho T. DPP4 and ACE2 in Diabetes and COVID-19: Therapeutic Targets for Cardiovascular Complications? Front Pharmacol 2020; 11:1161. [PMID: 32848769 PMCID: PMC7426477 DOI: 10.3389/fphar.2020.01161] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/17/2020] [Indexed: 12/15/2022] Open
Abstract
COVID-19 outbreak, caused by severe acute respiratory syndrome (SARS)-CoV-2 coronavirus has become an urgent health and economic challenge. Diabetes is a risk factor for severity and mortality of COVID-19. Recent studies support that COVID-19 has effects beyond the respiratory tract, with vascular complications arising as relevant factors worsening its prognosis, then making patients with previous vascular disease more prone to severity or fatal outcome. Angiotensin-II converting enzime-2 (ACE2) has been proposed as preferred receptor for SARS-CoV-2 host infection, yet specific proteins participating in the virus entry are not fully known. SARS-CoV-2 might use other co-receptor or auxiliary proteins allowing virus infection. In silico experiments proposed that SARS-CoV-2 might bind dipeptidyl peptidase 4 (DPP4/CD26), which was established previously as receptor for MERS-CoV. The renin-angiotensin-aldosterone system (RAAS) component ACE2 and DPP4 are proteins dysregulated in diabetes. Imbalance of the RAAS and direct effect of soluble DPP4 exert deleterious vascular effects. We hypothesize that diabetic patients might be more affected by COVID-19 due to increased presence ACE2 and DPP4 mediating infection and contributing to a compromised vasculature. Here, we discuss the role of ACE2 and DPP4 as relevant factors linking the risk of SARS-CoV-2 infection and severity of COVID-19 in diabetic patients and present an outlook on therapeutic potential of current drugs targeted against RAAS and DPP4 to treat or prevent COVID-19-derived vascular complications. Diabetes affects more than 400 million people worldwide, thus better understanding of how they are affected by COVID-19 holds an important benefit to fight against this disease with pandemic proportions.
Collapse
Affiliation(s)
- Inés Valencia
- Vascular Pharmacology and Metabolism Group (FARMAVASM), Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigaciones Sanitarias del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Concepción Peiró
- Vascular Pharmacology and Metabolism Group (FARMAVASM), Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigaciones Sanitarias del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Óscar Lorenzo
- Laboratory of Vascular Pathology and Diabetes, FIIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM) Network, Madrid, Spain
| | - Carlos F Sánchez-Ferrer
- Vascular Pharmacology and Metabolism Group (FARMAVASM), Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigaciones Sanitarias del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Jürgen Eckel
- German Diabetes Center, Institute for Clinical Diabetology, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Tania Romacho
- German Diabetes Center, Institute for Clinical Diabetology, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
37
|
CCR5-Δ32 gene variant frequency in the Turkish Cypriot population. Braz J Microbiol 2020; 51:1711-1717. [PMID: 32734471 PMCID: PMC7392619 DOI: 10.1007/s42770-020-00352-8] [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: 05/07/2020] [Accepted: 07/25/2020] [Indexed: 11/09/2022] Open
Abstract
Recent UNAIDS reports (December 2019) indicate that 37.9 million people have been affected by HIV infection around the globe in 2018, of which 1.7 million are cited as new infections. Human immunodeficiency virus-1 (HIV-1) requires both the CD4 receptor, as the primary receptor, and a chemokine co-receptor to gain entry into the cell. In addition to the WT allele for C–C motif chemokine receptor 5 (CCR5-wt), there is another allele with a 32 bp deletion in the protein coding region (CCR5-Δ32). Individuals who are homozygous for the mutant allele are resistant towards M-tropic HIV infections. In the current study, we aimed to determine the CCR5-Δ32 allele frequency in the Turkish Cypriot population with 326 subjects, 141 men (43.1%) and 185 (56.9%) women. The region of the CCR5 gene containing the Δ32 deletion was amplified using flanking primers. The CCR5 gene Δ32 allele frequency was calculated at 3% and only observed in heterozygous individuals. We hope that our current publication could be a point of dialog between the physicians, the government officials and the public set up a more modern and well-structured HIV screening program in an effort to control and hopefully eliminate HIV from the Turkish Cypriot population.
Collapse
|
38
|
García LF. Immune Response, Inflammation, and the Clinical Spectrum of COVID-19. Front Immunol 2020; 11:1441. [PMID: 32612615 PMCID: PMC7308593 DOI: 10.3389/fimmu.2020.01441] [Citation(s) in RCA: 452] [Impact Index Per Article: 113.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/03/2020] [Indexed: 01/08/2023] Open
Abstract
The current COVID-19 pandemic began in December 2019 in Wuhan (China) and rapidly extended to become a global sanitary and economic emergency. Its etiological agent is the coronavirus SARS-CoV-2. COVID-19 presents a wide spectrum of clinical manifestations, which ranges from an asymptomatic infection to a severe pneumonia accompanied by multisystemic failure that can lead to a patient's death. The immune response to SARS-CoV-2 is known to involve all the components of the immune system that together appear responsible for viral elimination and recovery from the infection. Nonetheless, such immune responses are implicated in the disease's progression to a more severe and lethal process. This review describes the general aspects of both COVID-19 and its etiological agent SARS-CoV-2, stressing the similarities with other severe coronavirus infections, such as SARS and MERS, but more importantly, pointing toward the evidence supporting the hypothesis that the clinical spectrum of COVID-19 is a consequence of the corresponding variable spectrum of the immune responses to the virus. The critical point where progression of the disease ensues appears to center on loss of the immune regulation between protective and altered responses due to exacerbation of the inflammatory components. Finally, it appears possible to delineate certain major challenges deserving of exhaustive investigation to further understand COVID-19 immunopathogenesis, thus helping to design more effective diagnostic, therapeutic, and prophylactic strategies.
Collapse
Affiliation(s)
- Luis F. García
- Grupo de Inmunología Celular e Inmunogenética, Facultad de Medicina, Sede de Investigación Universitaria, Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
39
|
Zohar T, Alter G. Dissecting antibody-mediated protection against SARS-CoV-2. Nat Rev Immunol 2020; 20:392-394. [PMID: 32514035 PMCID: PMC7278217 DOI: 10.1038/s41577-020-0359-5] [Citation(s) in RCA: 155] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2020] [Indexed: 12/18/2022]
Affiliation(s)
- Tomer Zohar
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
| |
Collapse
|
40
|
Hachim MY, Hachim IY, Naeem KB, Hannawi H, Al Salmi I, Hannawi S. C-C chemokine receptor type 5 links COVID-19, rheumatoid arthritis, and Hydroxychloroquine: in silico analysis. TRANSLATIONAL MEDICINE COMMUNICATIONS 2020; 5:14. [PMID: 32923679 PMCID: PMC7479747 DOI: 10.1186/s41231-020-00066-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 08/25/2020] [Indexed: 02/08/2023]
Abstract
Patients with rheumatoid arthritis (RA) represent one of the fragile patient groups that might be susceptible to the critical form of the coronavirus disease - 19 (COVID-19). On the other side, RA patients have been found not to have an increased risk of COVID-19 infection. Moreover, some of the Disease-Modifying Anti-Rheumatic Drugs (DMARDS) commonly used to treat rheumatic diseases like Hydroxychloroquine (HCQ) were proposed as a potential therapy for COVID-19 with a lack of full understanding of their molecular mechanisms. This highlights the need for the discovery of common pathways that may link both diseases at the molecular side. In this research, we used the in silico approach to investigate the transcriptomic profile of RA synovium to identify shared molecular pathways with that of severe acute respiratory syndrome-corona virus-2 (SARS-COV-2) infected lung tissue. Our results showed upregulation of chemotactic factors, including CCL4, CCL8, and CCL11, that all shared CCR5 as their receptor, as a common derangement observed in both diseases; RA and COVID-19. Moreover, our results also highlighted a possible mechanism through which HCQ, which can be used as a monotherapy in mild RA or as one of the triple-DMARDs therapy (tDMARDs; methotrexate, sulphasalazine, and HCQ), might interfere with the COVID-19 infection. This might be achieved through the ability of HCQ to upregulate specific immune cell populations like activated natural killer (NK) cells, which were found to be significantly reduced in COVID-19 infection. In addition to its ability to block CCR5 rich immune cell recruitment that also was upregulated in the SARS-COV-2 infected lungs. This might explain some of the reports that showed beneficial effects.
Collapse
Affiliation(s)
- Mahmood Y. Hachim
- College of Medicine, Mohammed bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Ibrahim Y. Hachim
- grid.412789.10000 0004 4686 5317Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, UAE
| | - Kashif Bin Naeem
- grid.415786.90000 0004 1773 3198Ministry of Health and Prevention (MOHAP), Dubai, UAE
| | - Haifa Hannawi
- grid.415786.90000 0004 1773 3198Ministry of Health and Prevention (MOHAP), Dubai, UAE
| | - Issa Al Salmi
- grid.416132.30000 0004 1772 5665The Royal Hospital, Muscat, Oman
| | - Suad Hannawi
- grid.415786.90000 0004 1773 3198Ministry of Health and Prevention (MOHAP), Dubai, UAE
| |
Collapse
|