1
|
Kovalenko A, Viatte S. Development and application of EpitopeScan, a Python3 toolset for mutation tracking in SARS-CoV-2 immunogenic epitopes. Front Immunol 2024; 15:1356314. [PMID: 38840924 PMCID: PMC11150570 DOI: 10.3389/fimmu.2024.1356314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 05/06/2024] [Indexed: 06/07/2024] Open
Abstract
Introduction Outbreaks of coronaviruses and especially the recent COVID-19 pandemic emphasize the importance of immunological research in this area to mitigate the effect of future incidents. Bioinformatics approaches are capable of providing multisided insights from virus sequencing data, although currently available software options are not entirely suitable for a specific task of mutation surveillance within immunogenic epitopes of SARS-CoV-2. Method Here, we describe the development of a mutation tracker, EpitopeScan, a Python3 package with command line and graphical user interface tools facilitating the investigation of the mutation dynamics in SARS-CoV-2 epitopes via analysis of multiple-sequence alignments of genomes over time. We provide an application case by examining three Spike protein-derived immunodominant CD4+ T-cell epitopes restricted by HLA-DRB1*04:01, an allele strongly associated with susceptibility to rheumatoid arthritis (RA). Mutations in these peptides are relevant for immune monitoring of CD4+ T-cell responses against SARS-CoV-2 spike protein in patients with RA. The analysis focused on 2.3 million SARS-CoV-2 genomes sampled in England. Results We detail cases of epitope conservation over time, partial loss of conservation, and complete divergence from the wild type following the emergence of the N969K Omicron-specific mutation in November 2021. The wild type and the mutated peptide represent potential candidates to monitor variant-specific CD4+ T-cell responses. EpitopeScan is available via GitHub repository https://github.com/Aleksandr-biochem/EpitopeScan.
Collapse
Affiliation(s)
- Alexander Kovalenko
- Versus Arthritis Centre for Genetics and Genomics, Centre for Musculoskeletal Research, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| | - Sebastien Viatte
- Versus Arthritis Centre for Genetics and Genomics, Centre for Musculoskeletal Research, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- National Institute for Health and Care Research (NIHR) Manchester Musculoskeletal Biomedical Research Centre, Central Manchester National Health Service (NHS) Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|
2
|
Duette G, Lee E, Martins Costa Gomes G, Tungatt K, Doyle C, Stylianou VV, Lee A, Maddocks S, Taylor J, Khanna R, Bull RA, Martinello M, Sandgren KJ, Cunningham AL, Palmer S. Highly Networked SARS-CoV-2 Peptides Elicit T Cell Responses with Enhanced Specificity. Immunohorizons 2023; 7:508-527. [PMID: 37358499 PMCID: PMC10580120 DOI: 10.4049/immunohorizons.2300034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 06/05/2023] [Indexed: 06/27/2023] Open
Abstract
Identifying SARS-CoV-2-specific T cell epitope-derived peptides is critical for the development of effective vaccines and measuring the duration of specific SARS-CoV-2 cellular immunity. In this regard, we previously identified T cell epitope-derived peptides within topologically and structurally essential regions of SARS-CoV-2 spike and nucleocapsid proteins by applying an immunoinformatics pipeline. In this study, we selected 30 spike- and nucleocapsid-derived peptides and assessed whether these peptides induce T cell responses and avoid major mutations found in SARS-CoV-2 variants of concern. Our peptide pool was highly specific, with only a single peptide driving cross-reactivity in people unexposed to SARS-COV-2, and immunogenic, inducing a polyfunctional response in CD4+ and CD8+ T cells from COVID-19 recovered individuals. All peptides were immunogenic and individuals recognized broad and diverse peptide repertoires. Moreover, our peptides avoided most mutations/deletions associated with all four SARS-CoV-2 variants of concern while retaining their physicochemical properties even when genetic changes are introduced. This study contributes to an evolving definition of individual CD4+ and CD8+ T cell epitopes that can be used for specific diagnostic tools for SARS-CoV-2 T cell responses and is relevant to the development of variant-resistant and durable T cell-stimulating vaccines.
Collapse
Affiliation(s)
- Gabriel Duette
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Eunok Lee
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | | | - Katie Tungatt
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Chloe Doyle
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Vicki V. Stylianou
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Ashley Lee
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Susan Maddocks
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, New South Wales, Australia
| | - Janette Taylor
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, New South Wales, Australia
| | - Rajiv Khanna
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Rowena A. Bull
- School of Medical Sciences, Faculty of Medicine, UNSW Australia, Sydney, New South Wales, Australia
- The Kirby Institute, UNSW Australia, Sydney, New South Wales, Australia
| | - Marianne Martinello
- The Kirby Institute, UNSW Australia, Sydney, New South Wales, Australia
- Westmead Hospital, Sydney, New South Wales, Australia
- Blacktown & Mount Druitt Hospital, Blacktown, New South Wales, Australia
| | - Kerrie J. Sandgren
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Anthony L. Cunningham
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Sarah Palmer
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
3
|
Ghafouri F, Ahangari Cohan R, Samimi H, Hosseini Rad S M A, Naderi M, Noorbakhsh F, Haghpanah V. Development of a Multiepitope Vaccine Against SARS-CoV-2: Immunoinformatics Study. JMIR BIOINFORMATICS AND BIOTECHNOLOGY 2022; 3:e36100. [PMID: 35891920 PMCID: PMC9302570 DOI: 10.2196/36100] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 05/16/2022] [Accepted: 07/04/2022] [Indexed: 11/21/2022]
Abstract
Background Since the first appearance of SARS-CoV-2 in China in December 2019, the world witnessed the emergence of the SARS-CoV-2 outbreak. Due to the high transmissibility rate of the virus, there is an urgent need to design and develop vaccines against SARS-CoV-2 to prevent more cases affected by the virus. Objective A computational approach is proposed for vaccine design against the SARS-CoV-2 spike (S) protein, as the key target for neutralizing antibodies, and envelope (E) protein, which contains a conserved sequence feature. Methods We used previously reported epitopes of S protein detected experimentally and further identified a collection of predicted B-cell and major histocompatibility (MHC) class II–restricted T-cell epitopes derived from E proteins with an identical match to SARS-CoV-2 E protein. Results The in silico design of our candidate vaccine against the S and E proteins of SARS-CoV-2 demonstrated a high affinity to MHC class II molecules and effective results in immune response simulations. Conclusions Based on the results of this study, the multiepitope vaccine designed against the S and E proteins of SARS-CoV-2 may be considered as a new, safe, and efficient approach to combatting the COVID-19 pandemic.
Collapse
Affiliation(s)
- Fatemeh Ghafouri
- Department of Biotechnology Faculty of Life Sciences and Biotechnology Shahid Beheshti University Tehran Iran
| | - Reza Ahangari Cohan
- Department of Nanobiotechnology New Technologies Research Group Pasteur Institute of Iran Tehran Iran
| | - Hilda Samimi
- Endocrinology and Metabolism Research Center Endocrinology and Metabolism Clinical Sciences Institute Tehran University of Medical Sciences Tehran Iran
| | | | - Mahmood Naderi
- Digestive Diseases Research Center Digestive Diseases Research Institute Tehran University of Medical Sciences Tehran Iran
| | - Farshid Noorbakhsh
- Department of Immunology School of Medicine Tehran University of Medical Sciences Tehran Iran
| | - Vahid Haghpanah
- Endocrinology and Metabolism Research Center Endocrinology and Metabolism Clinical Sciences Institute Tehran University of Medical Sciences Tehran Iran
- Personalized Medicine Research Center Endocrinology and Metabolism Clinical Sciences Institute Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
4
|
Becerra-Artiles A, Calvo-Calle JM, Co MD, Nanaware PP, Cruz J, Weaver GC, Lu L, Forconi C, Finberg RW, Moormann AM, Stern LJ. Broadly recognized, cross-reactive SARS-CoV-2 CD4 T cell epitopes are highly conserved across human coronaviruses and presented by common HLA alleles. Cell Rep 2022; 39:110952. [PMID: 35675811 PMCID: PMC9135679 DOI: 10.1016/j.celrep.2022.110952] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 04/03/2022] [Accepted: 05/23/2022] [Indexed: 11/24/2022] Open
Abstract
Sequence homology between SARS-CoV-2 and common-cold human coronaviruses (HCoVs) raises the possibility that memory responses to prior HCoV infection can affect T cell response in COVID-19. We studied T cell responses to SARS-CoV-2 and HCoVs in convalescent COVID-19 donors and identified a highly conserved SARS-CoV-2 sequence, S811-831, with overlapping epitopes presented by common MHC class II proteins HLA-DQ5 and HLA-DP4. These epitopes are recognized by low-abundance CD4 T cells from convalescent COVID-19 donors, mRNA vaccine recipients, and uninfected donors. TCR sequencing revealed a diverse repertoire with public TCRs. T cell cross-reactivity is driven by the high conservation across human and animal coronaviruses of T cell contact residues in both HLA-DQ5 and HLA-DP4 binding frames, with distinct patterns of HCoV cross-reactivity explained by MHC class II binding preferences and substitutions at secondary TCR contact sites. These data highlight S811-831 as a highly conserved CD4 T cell epitope broadly recognized across human populations.
Collapse
Affiliation(s)
| | | | - Mary Dawn Co
- Department of Medicine, UMass Chan Medical School, Worcester, MA 01655, USA
| | - Padma P Nanaware
- Department of Pathology, UMass Chan Medical School, Worcester, MA 01655, USA
| | - John Cruz
- Department of Pathology, UMass Chan Medical School, Worcester, MA 01655, USA
| | - Grant C Weaver
- Department of Pathology, UMass Chan Medical School, Worcester, MA 01655, USA
| | - Liying Lu
- Department of Pathology, UMass Chan Medical School, Worcester, MA 01655, USA
| | - Catherine Forconi
- Department of Medicine, UMass Chan Medical School, Worcester, MA 01655, USA
| | - Robert W Finberg
- Department of Medicine, UMass Chan Medical School, Worcester, MA 01655, USA
| | - Ann M Moormann
- Department of Medicine, UMass Chan Medical School, Worcester, MA 01655, USA
| | - Lawrence J Stern
- Department of Pathology, UMass Chan Medical School, Worcester, MA 01655, USA; Department of Biochemistry and Molecular Biotechnology, UMass Chan Medical School, Worcester, MA 01655, USA.
| |
Collapse
|
5
|
Fahrner JE, Lahmar I, Goubet AG, Haddad Y, Carrier A, Mazzenga M, Drubay D, Alves Costa Silva C, Lyon COVID Study Group, de Sousa E, Thelemaque C, Melenotte C, Dubuisson A, Geraud A, Ferrere G, Birebent R, Bigenwald C, Picard M, Cerbone L, Lérias JR, Laparra A, Bernard-Tessier A, Kloeckner B, Gazzano M, Danlos FX, Terrisse S, Pizzato E, Flament C, Ly P, Tartour E, Benhamouda N, Meziani L, Ahmed-Belkacem A, Miyara M, Gorochov G, Barlesi F, Trubert A, Ungar B, Estrada Y, Pradon C, Gallois E, Pommeret F, Colomba E, Lavaud P, Deloger M, Droin N, Deutsch E, Gachot B, Spano JP, Merad M, Scotté F, Marabelle A, Griscelli F, Blay JY, Soria JC, Merad M, André F, Villemonteix J, Chevalier MF, Caillat-Zucman S, Fenollar F, Guttman-Yassky E, Launay O, Kroemer G, La Scola B, Maeurer M, Derosa L, Zitvogel L. The Polarity and Specificity of Antiviral T Lymphocyte Responses Determine Susceptibility to SARS-CoV-2 Infection in Patients with Cancer and Healthy Individuals. Cancer Discov 2022; 12:958-983. [PMID: 35179201 PMCID: PMC9394394 DOI: 10.1158/2159-8290.cd-21-1441] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/10/2022] [Accepted: 01/31/2022] [Indexed: 01/07/2023]
Abstract
Vaccination against coronavirus disease 2019 (COVID-19) relies on the in-depth understanding of protective immune responses to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). We characterized the polarity and specificity of memory T cells directed against SARS-CoV-2 viral lysates and peptides to determine correlates with spontaneous, virus-elicited, or vaccine-induced protection against COVID-19 in disease-free and cancer-bearing individuals. A disbalance between type 1 and 2 cytokine release was associated with high susceptibility to COVID-19. Individuals susceptible to infection exhibited a specific deficit in the T helper 1/T cytotoxic 1 (Th1/Tc1) peptide repertoire affecting the receptor binding domain of the spike protein (S1-RBD), a hotspot of viral mutations. Current vaccines triggered Th1/Tc1 responses in only a fraction of all subject categories, more effectively against the original sequence of S1-RBD than that from viral variants. We speculate that the next generation of vaccines should elicit Th1/Tc1 T-cell responses against the S1-RBD domain of emerging viral variants. SIGNIFICANCE This study prospectively analyzed virus-specific T-cell correlates of protection against COVID-19 in healthy and cancer-bearing individuals. A disbalance between Th1/Th2 recall responses conferred susceptibility to COVID-19 in both populations, coinciding with selective defects in Th1 recognition of the receptor binding domain of spike. See related commentary by McGary and Vardhana, p. 892. This article is highlighted in the In This Issue feature, p. 873.
Collapse
Affiliation(s)
- Jean-Eudes Fahrner
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France.,Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France.,Transgene S.A., Illkirch-Graffenstaden, France
| | - Imran Lahmar
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France.,Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France
| | - Anne-Gaëlle Goubet
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France.,Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France
| | - Yacine Haddad
- Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France
| | - Agathe Carrier
- Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France
| | - Marine Mazzenga
- Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France
| | - Damien Drubay
- Gustave Roussy, Villejuif, France.,Département de Biostatistique et d'Epidémiologie, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Carolina Alves Costa Silva
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France.,Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France
| | - Lyon COVID Study Group
- Open Innovation & Partnerships (OIP), bioMérieux S.A., Marcy l'Etoile, France. R&D – Immunoassay, bioMérieux S.A., Marcy l'Etoile, France.,Joint Research Unit Hospices Civils de Lyon-bioMérieux, Civils Hospices of Lyon, Lyon Sud Hospital, Pierre-Bénite, France.,International Center of Research in Infectiology, Lyon University, INSERM U1111, CNRS UMR 5308, ENS, UCBL, Lyon, France.,Hospices Civils de Lyon, Lyon Sud Hospital, Pierre-Bénite, France
| | - Eric de Sousa
- ImmunoTherapy/ImmunoSurgery, Champalimaud Centre for the Unknown, Lisboa, Portugal
| | - Cassandra Thelemaque
- Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France
| | - Cléa Melenotte
- Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France.,Aix-Marseille Université, Institut Hospitalo-Universitaire, Institut de Recherche pour le Développement, Assistance Publique – Hôpitaux de Marseille, Microbes Evolution Phylogeny and Infections, Marseille, France
| | - Agathe Dubuisson
- Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France
| | - Arthur Geraud
- Gustave Roussy, Villejuif, France.,Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Villejuif, France.,Département d'Oncologie Médicale, Gustave Roussy, Villejuif, France
| | - Gladys Ferrere
- Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France
| | - Roxanne Birebent
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France.,Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France
| | - Camille Bigenwald
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France.,Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France
| | - Marion Picard
- Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France
| | - Luigi Cerbone
- Gustave Roussy, Villejuif, France.,Département d'Oncologie Médicale, Gustave Roussy, Villejuif, France
| | - Joana R. Lérias
- ImmunoTherapy/ImmunoSurgery, Champalimaud Centre for the Unknown, Lisboa, Portugal
| | - Ariane Laparra
- Gustave Roussy, Villejuif, France.,Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Villejuif, France.,Département d'Oncologie Médicale, Gustave Roussy, Villejuif, France
| | - Alice Bernard-Tessier
- Gustave Roussy, Villejuif, France.,Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Villejuif, France.,Département d'Oncologie Médicale, Gustave Roussy, Villejuif, France
| | - Benoît Kloeckner
- Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France
| | - Marianne Gazzano
- Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France
| | - François-Xavier Danlos
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France.,Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France
| | - Safae Terrisse
- Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France
| | - Eugenie Pizzato
- Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France
| | - Caroline Flament
- Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France
| | - Pierre Ly
- Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France
| | - Eric Tartour
- Center of clinical investigations BIOTHERIS, INSERM CIC1428, Gustave Roussy, Villejuif, France.,Department of Immunology, Hôpital Européen Georges Pompidou, APHP, Paris, France
| | - Nadine Benhamouda
- Center of clinical investigations BIOTHERIS, INSERM CIC1428, Gustave Roussy, Villejuif, France.,Department of Immunology, Hôpital Européen Georges Pompidou, APHP, Paris, France
| | | | | | - Makoto Miyara
- Univ Paris Est Créteil, INSERM U955, IMRB, Créteil, France
| | - Guy Gorochov
- Univ Paris Est Créteil, INSERM U955, IMRB, Créteil, France
| | - Fabrice Barlesi
- Gustave Roussy, Villejuif, France.,Département d'Oncologie Médicale, Gustave Roussy, Villejuif, France.,Sorbonne Université/Institut National de la Santé et de la Recherche Médicale, U1135, Centre d'Immunologie et des Maladies Infectieuses, Hôpital Pitié-Salpêtrière, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Alexandre Trubert
- Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France
| | - Benjamin Ungar
- Aix Marseille University, CNRS, INSERM, CRCM, Marseille, France
| | - Yeriel Estrada
- Department of Dermatology, Center of Excellence in Eczema Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Caroline Pradon
- Gustave Roussy, Villejuif, France.,Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York.,Centre de Ressources Biologiques, ET-EXTRA, Gustave Roussy, Villejuif, France
| | - Emmanuelle Gallois
- Gustave Roussy, Villejuif, France.,Département de Biologie Médicale et Pathologie Médicales, Service de Biochimie, Gustave Roussy, Villejuif, France
| | - Fanny Pommeret
- Gustave Roussy, Villejuif, France.,Département d'Oncologie Médicale, Gustave Roussy, Villejuif, France
| | - Emeline Colomba
- Gustave Roussy, Villejuif, France.,Département d'Oncologie Médicale, Gustave Roussy, Villejuif, France
| | - Pernelle Lavaud
- Gustave Roussy, Villejuif, France.,Département d'Oncologie Médicale, Gustave Roussy, Villejuif, France
| | - Marc Deloger
- Département de Biologie Médicale et Pathologie Médicales, Service de Microbiologie, Gustave Roussy, Villejuif, France
| | - Nathalie Droin
- Gustave Roussy, Plateforme de Bioinformatique, Université Paris-Saclay, INSERM US23, CNRS UMS, Villejuif, France
| | - Eric Deutsch
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France.,Gustave Roussy, Villejuif, France.,Gustave Roussy, Plateforme de génomique, Université Paris-Saclay, INSERM US23, CNRS UMS, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, U1030, Gustave Roussy, Villejuif, France
| | - Bertrand Gachot
- Gustave Roussy, Villejuif, France.,Département de Radiothérapie, Gustave Roussy, Villejuif, France
| | | | - Mansouria Merad
- Gustave Roussy, Villejuif, France.,Department of Medical Oncology, Pitié-Salpétrière Hospital, APHP, Sorbonne Université, Paris, France
| | - Florian Scotté
- Gustave Roussy, Villejuif, France.,Service de Médecine aigue d’Urgence en Cancérologie, Gustave Roussy, Villejuif, France
| | - Aurélien Marabelle
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France.,Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France.,Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Villejuif, France.,Département d'Oncologie Médicale, Gustave Roussy, Villejuif, France.,Département Interdisciplinaire d'Organisation des Parcours Patients, Gustave Roussy, Villejuif, France
| | - Frank Griscelli
- Gustave Roussy, Villejuif, France.,Département de Biologie Médicale et Pathologie Médicales, Service de Biochimie, Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale – UMR935/UA9, Université Paris-Saclay, Villejuif, France.,INGESTEM National IPSC Infrastructure, Université de Paris-Saclay, Villejuif, France.,Université de Paris, Faculté des Sciences Pharmaceutiques et Biologiques, Paris, France
| | - Jean-Yves Blay
- Centre Léon Bérard, Lyon, France.,Université Claude Bernard, Lyon, France.,Unicancer, Paris, France
| | - Jean-Charles Soria
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France.,Gustave Roussy, Villejuif, France
| | - Miriam Merad
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Oncological Science, Icahn School of Medicine at Mount Sinai, New York, New York.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Fabrice André
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France.,Gustave Roussy, Villejuif, France.,Département d'Oncologie Médicale, Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, U981, Gustave Roussy, Villejuif, France
| | - Juliette Villemonteix
- Laboratoire d'Immunologie et Histocompatibilité, Hôpital Saint-Louis, APHP, Université de Paris, Paris, France
| | - Mathieu F. Chevalier
- INSERM UMR 976, Institut de Recherche Saint-Louis, Université de Paris, Paris, France
| | - Sophie Caillat-Zucman
- Laboratoire d'Immunologie et Histocompatibilité, Hôpital Saint-Louis, APHP, Université de Paris, Paris, France.,INSERM UMR 976, Institut de Recherche Saint-Louis, Université de Paris, Paris, France
| | - Florence Fenollar
- IHU Méditérranée Infection, VITROME, IRD, AP-HM, SSA, Aix-Marseille University, Marseille, France
| | - Emma Guttman-Yassky
- Department of Dermatology, Center of Excellence in Eczema Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Odile Launay
- Université de Paris, Inserm CIC 1417, I-Reivac, APHP, Hopital Cochin, Paris, France
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif, France.,Pôle de Biologie, Hôpital Européen George Pompidou, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Bernard La Scola
- Institut Hospitalo-Universitaire, Méditerranée Infection, Marseille, France
| | - Markus Maeurer
- ImmunoTherapy/ImmunoSurgery, Champalimaud Centre for the Unknown, Lisboa, Portugal.,Medizinische Klinik, Johannes Gutenberg University Mainz, Germany
| | - Lisa Derosa
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France.,Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France.,Département d'Oncologie Médicale, Gustave Roussy, Villejuif, France
| | - Laurence Zitvogel
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France.,Gustave Roussy, Villejuif, France.,Institut National de la Santé et de la Recherche Médicale, UMR1015, Gustave Roussy, Villejuif, France.,Center of clinical investigations BIOTHERIS, INSERM CIC1428, Gustave Roussy, Villejuif, France.,Corresponding Author: Laurence Zitvogel, University Paris-Saclay, Gustave Roussy Cancer Center, 114 rue Edouard Vaillant, Villejuif Cedex 94805, France. Phone: 331-4211-5041; E-mail:
| |
Collapse
|
6
|
Liu R, Jiang W, Mellins ED. Yeast display of MHC-II enables rapid identification of peptide ligands from protein antigens (RIPPA). Cell Mol Immunol 2021; 18:1847-1860. [PMID: 34117370 PMCID: PMC8193015 DOI: 10.1038/s41423-021-00717-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 05/25/2021] [Indexed: 11/12/2022] Open
Abstract
CD4+ T cells orchestrate adaptive immune responses via binding of antigens to their receptors through specific peptide/MHC-II complexes. To study these responses, it is essential to identify protein-derived MHC-II peptide ligands that constitute epitopes for T cell recognition. However, generating cells expressing single MHC-II alleles and isolating these proteins for use in peptide elution or binding studies is time consuming. Here, we express human MHC alleles (HLA-DR4 and HLA-DQ6) as native, noncovalent αβ dimers on yeast cells for direct flow cytometry-based screening of peptide ligands from selected antigens. We demonstrate rapid, accurate identification of DQ6 ligands from pre-pro-hypocretin, a narcolepsy-related immunogenic target. We also identify 20 DR4-binding SARS-CoV-2 spike peptides homologous to SARS-CoV-1 epitopes, and one spike peptide overlapping with the reported SARS-CoV-2 epitope recognized by CD4+ T cells from unexposed individuals carrying DR4 subtypes. Our method is optimized for immediate application upon the emergence of novel pathogens.
Collapse
Affiliation(s)
- Rongzeng Liu
- Department of Pediatrics-Human Gene Therapy, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Immunology, Henan University of Science and Technology School of Medicine, Luoyang, China
| | - Wei Jiang
- Department of Pediatrics-Human Gene Therapy, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Immunology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Elizabeth D Mellins
- Department of Pediatrics-Human Gene Therapy, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Immunology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
7
|
Smith CC, Olsen KS, Gentry KM, Sambade M, Beck W, Garness J, Entwistle S, Willis C, Vensko S, Woods A, Fini M, Carpenter B, Routh E, Kodysh J, O'Donnell T, Haber C, Heiss K, Stadler V, Garrison E, Sandor AM, Ting JPY, Weiss J, Krajewski K, Grant OC, Woods RJ, Heise M, Vincent BG, Rubinsteyn A. Landscape and selection of vaccine epitopes in SARS-CoV-2. Genome Med 2021; 13:101. [PMID: 34127050 PMCID: PMC8201469 DOI: 10.1186/s13073-021-00910-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 05/14/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Early in the pandemic, we designed a SARS-CoV-2 peptide vaccine containing epitope regions optimized for concurrent B cell, CD4+ T cell, and CD8+ T cell stimulation. The rationale for this design was to drive both humoral and cellular immunity with high specificity while avoiding undesired effects such as antibody-dependent enhancement (ADE). METHODS We explored the set of computationally predicted SARS-CoV-2 HLA-I and HLA-II ligands, examining protein source, concurrent human/murine coverage, and population coverage. Beyond MHC affinity, T cell vaccine candidates were further refined by predicted immunogenicity, sequence conservation, source protein abundance, and coverage of high frequency HLA alleles. B cell epitope regions were chosen from linear epitope mapping studies of convalescent patient serum, followed by filtering for surface accessibility, sequence conservation, spatial localization near functional domains of the spike glycoprotein, and avoidance of glycosylation sites. RESULTS From 58 initial candidates, three B cell epitope regions were identified. From 3730 (MHC-I) and 5045 (MHC-II) candidate ligands, 292 CD8+ and 284 CD4+ T cell epitopes were identified. By combining these B cell and T cell analyses, as well as a manufacturability heuristic, we proposed a set of 22 SARS-CoV-2 vaccine peptides for use in subsequent murine studies. We curated a dataset of ~ 1000 observed T cell epitopes from convalescent COVID-19 patients across eight studies, showing 8/15 recurrent epitope regions to overlap with at least one of our candidate peptides. Of the 22 candidate vaccine peptides, 16 (n = 10 T cell epitope optimized; n = 6 B cell epitope optimized) were manually selected to decrease their degree of sequence overlap and then synthesized. The immunogenicity of the synthesized vaccine peptides was validated using ELISpot and ELISA following murine vaccination. Strong T cell responses were observed in 7/10 T cell epitope optimized peptides following vaccination. Humoral responses were deficient, likely due to the unrestricted conformational space inhabited by linear vaccine peptides. CONCLUSIONS Overall, we find our selection process and vaccine formulation to be appropriate for identifying T cell epitopes and eliciting T cell responses against those epitopes. Further studies are needed to optimize prediction and induction of B cell responses, as well as study the protective capacity of predicted T and B cell epitopes.
Collapse
Affiliation(s)
- Christof C Smith
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Kelly S Olsen
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Kaylee M Gentry
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Maria Sambade
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Wolfgang Beck
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Jason Garness
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Sarah Entwistle
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Caryn Willis
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Steven Vensko
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Allison Woods
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, NC, USA
| | - Misha Fini
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, NC, USA
| | - Brandon Carpenter
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Eric Routh
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Julia Kodysh
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Timothy O'Donnell
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | - Erik Garrison
- Genomics Institute, University of California, Santa Cruz, CA, USA
| | - Adam M Sandor
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
| | - Jenny P Y Ting
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
- Department of Genetics, UNC School of Medicine, Chapel Hill, NC, USA
- Institute for Inflammatory Diseases, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Center for Translational Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jared Weiss
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
- Division of Medical Oncology, Department of Medicine, UNC School of Medicine, Chapel Hill, NC, USA
| | - Krzysztof Krajewski
- Department of Biochemistry and Biophysics, UNC School of Medicine, Chapel Hill, NC, USA
| | - Oliver C Grant
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Robert J Woods
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Mark Heise
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA
- Department of Genetics, UNC School of Medicine, Chapel Hill, NC, USA
| | - Benjamin G Vincent
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, NC, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA.
- Computational Medicine Program, UNC School of Medicine, Chapel Hill, NC, USA.
- Curriculum in Bioinformatics and Computational Biology, UNC School of Medicine, Chapel Hill, NC, USA.
- Division of Hematology, Department of Medicine, UNC School of Medicine, Chapel Hill, NC, USA.
| | - Alex Rubinsteyn
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, CB# 7295, Chapel Hill, NC, 27599-7295, USA.
- Department of Genetics, UNC School of Medicine, Chapel Hill, NC, USA.
- Computational Medicine Program, UNC School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
8
|
Mapping the SARS-CoV-2 spike glycoprotein-derived peptidome presented by HLA class II on dendritic cells. Cell Rep 2021; 35:109179. [PMID: 34004174 PMCID: PMC8116342 DOI: 10.1016/j.celrep.2021.109179] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 02/16/2021] [Accepted: 05/04/2021] [Indexed: 12/15/2022] Open
Abstract
Understanding and eliciting protective immune responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an urgent priority. To facilitate these objectives, we profile the repertoire of human leukocyte antigen class II (HLA-II)-bound peptides presented by HLA-DR diverse monocyte-derived dendritic cells pulsed with SARS-CoV-2 spike (S) protein. We identify 209 unique HLA-II-bound peptide sequences, many forming nested sets, which map to sites throughout S including glycosylated regions. Comparison of the glycosylation profile of the S protein to that of the HLA-II-bound S peptides reveals substantial trimming of glycan residues on the latter, likely induced during antigen processing. Our data also highlight the receptor-binding motif in S1 as a HLA-DR-binding peptide-rich region and identify S2-derived peptides with potential for targeting by cross-protective vaccine-elicited responses. Results from this study will aid analysis of CD4+ T cell responses in infected individuals and vaccine recipients and have application in next-generation vaccine design.
Collapse
|
9
|
Potential Antiviral Immune Response Against COVID-19: Lessons Learned from SARS-CoV. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1318:149-167. [PMID: 33973177 DOI: 10.1007/978-3-030-63761-3_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Virus and host innate immune system interaction plays a significant role in forming the outcome of viral diseases. Host innate immunity initially recognizes the viral invasion and induces a rapid inflammatory response, and this recognition activates signaling cascades that trigger the release of antiviral mediators. This chapter aims to explore the mechanisms by which newly emerged coronavirus called severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) activates the host immune system. Since SARS-CoV-2 shares similarities with SARS-CoV that caused the epidemic of SARS in 2003, the pathogenesis of both viruses could be at least very similar. For this, this chapter provides a synthesis of literature concerning antiviral immunity in SARS-CoV and SARS-CoV-2. It includes the presentation of epitopes linked to SARS-CoV-2 as well as the ability of SARS-CoV-2 to cause proteolytic activation and interact with angiotensin-converting enzyme 2 (ACE2) via molecular mimicry. This chapter characterizes various mechanisms that this virus may engage in escaping the host immunity, ended by a discussion of humoral immune responses against SARS-CoV-2.
Collapse
|
10
|
Alam A, Khan A, Imam N, Siddiqui MF, Waseem M, Malik MZ, Ishrat R. Design of an epitope-based peptide vaccine against the SARS-CoV-2: a vaccine-informatics approach. Brief Bioinform 2021; 22:1309-1323. [PMID: 33285567 PMCID: PMC7799329 DOI: 10.1093/bib/bbaa340] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/24/2020] [Accepted: 10/27/2020] [Indexed: 12/11/2022] Open
Abstract
The recurrent and recent global outbreak of the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has turned into a global concern which has infected more than 42 million people all over the globe, and this number is increasing in hours. Unfortunately, no vaccine or specific treatment is available, which makes it more deadly. A vaccine-informatics approach has shown significant breakthrough in peptide-based epitope mapping and opens the new horizon in vaccine development. In this study, we have identified a total of 15 antigenic peptides [including thymus cells (T-cells) and bone marrow or bursa-derived cells] in the surface glycoprotein (SG) of SARS-CoV-2 which is nontoxic and nonallergenic in nature, nonallergenic, highly antigenic and non-mutated in other SARS-CoV-2 virus strains. The population coverage analysis has found that cluster of differentiation 4 (CD4+) T-cell peptides showed higher cumulative population coverage over cluster of differentiation 8 (CD8+) peptides in the 16 different geographical regions of the world. We identified 12 peptides ((LTDEMIAQY, WTAGAAAYY, WMESEFRVY, IRASANLAA, FGAISSVLN, VKQLSSNFG, FAMQMAYRF, FGAGAALQI, YGFQPTNGVGYQ, LPDPSKPSKR, QTQTNSPRRARS and VITPGTNTSN) that are $80\hbox{--} 90\%$ identical with experimentally determined epitopes of SARS-CoV, and this will likely be beneficial for a quick progression of the vaccine design. Moreover, docking analysis suggested that the identified peptides are tightly bound in the groove of human leukocyte antigen molecules which can induce the T-cell response. Overall, this study allows us to determine potent peptide antigen targets in the SG on intuitive grounds, which opens up a new horizon in the coronavirus disease (COVID-19) research. However, this study needs experimental validation by in vitro and in vivo.
Collapse
Affiliation(s)
- Aftab Alam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia University, New Delhi 110025, India
| | - Arbaaz Khan
- Department of computer science, Jamia Millia Islamia University, New Delhi, India
| | - Nikhat Imam
- Centre for Interdisciplinary Research in Basic Science, Jamia Millia Islamia University, New Delhi, India
| | | | - Mohd Waseem
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Md Zubbair Malik
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Romana Ishrat
- Centre for Interdisciplinary Research in Basic Science, Jamia Millia Islamia University, New Delhi, India
| |
Collapse
|
11
|
Knierman MD, Lannan MB, Spindler LJ, McMillian CL, Konrad RJ, Siegel RW. The Human Leukocyte Antigen Class II Immunopeptidome of the SARS-CoV-2 Spike Glycoprotein. Cell Rep 2020; 33:108454. [PMID: 33220791 PMCID: PMC7664343 DOI: 10.1016/j.celrep.2020.108454] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/16/2020] [Accepted: 11/06/2020] [Indexed: 12/20/2022] Open
Abstract
Precise elucidation of the antigen sequences for T cell immunosurveillance greatly enhances our ability to understand and modulate humoral responses to viral infection or active immunization. Mass spectrometry is used to identify 526 unique sequences from the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike glycoprotein extracellular domain in a complex with human leukocyte antigen class II molecules on antigen-presenting cells from a panel of healthy donors selected to represent a majority of allele usage from this highly polymorphic molecule. The identified sequences span the entire spike protein, and several sequences are isolated from a majority of the sampled donors, indicating promiscuous binding. Importantly, many peptides derived from the receptor binding domain used for cell entry are identified. This work represents a precise and comprehensive immunopeptidomic investigation with the SARS-CoV-2 spike glycoprotein and allows detailed analysis of features that may aid vaccine development to end the current coronavirus disease 2019 (COVID-19) pandemic.
Collapse
Affiliation(s)
- Michael D Knierman
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Megan B Lannan
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Laura J Spindler
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Carl L McMillian
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Robert J Konrad
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Robert W Siegel
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA.
| |
Collapse
|
12
|
Liu T, Luo S, Libby P, Shi GP. Cathepsin L-selective inhibitors: A potentially promising treatment for COVID-19 patients. Pharmacol Ther 2020; 213:107587. [PMID: 32470470 PMCID: PMC7255230 DOI: 10.1016/j.pharmthera.2020.107587] [Citation(s) in RCA: 204] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2020] [Indexed: 12/19/2022]
Abstract
The widespread coronavirus SARS-CoV-2 has already infected over 4 million people worldwide, with a death toll over 280,000. Current treatment of COVID-19 patients relies mainly on antiviral drugs lopinavir/ritonavir, arbidol, and remdesivir, the anti-malarial drugs hydroxychloroquine and chloroquine, and traditional Chinese medicine. There are over 2,118 on-going clinical trials underway, but to date none of these drugs have consistently proven effective. Cathepsin L (CatL) is an endosomal cysteine protease. It mediates the cleavage of the S1 subunit of the coronavirus surface spike glycoprotein. This cleavage is necessary for coronavirus entry into human host cells, virus and host cell endosome membrane fusion, and viral RNA release for next round of replication. Here we summarize data regarding seven CatL-selective inhibitors that block coronavirus entry into cultured host cells and provide a mechanism to block SARS-CoV-2 infection in humans. Given the rapid growth of the SARS-CoV-2-positive population worldwide, ready-to-use CatL inhibitors should be explored as a treatment option. We identify ten US FDA-approved drugs that have CatL inhibitory activity. We provide evidence that supports the combined use of serine protease and CatL inhibitors as a possibly safer and more effective therapy than other available therapeutics to block coronavirus host cell entry and intracellular replication, without compromising the immune system.
Collapse
Affiliation(s)
- Tianxiao Liu
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Songyuan Luo
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Peter Libby
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
13
|
Parker R, Partridge T, Wormald C, Kawahara R, Stalls V, Aggelakopoulou M, Parker J, Doherty RP, Morejon YA, Lee E, Saunders K, Haynes BF, Acharya P, Thaysen-Andersen M, Borrow P, Ternette N. Mapping the SARS-CoV-2 spike glycoprotein-derived peptidome presented by HLA class II on dendritic cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 32839772 PMCID: PMC7444283 DOI: 10.1101/2020.08.19.255901] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Understanding and eliciting protective immune responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an urgent priority. To facilitate these objectives, we have profiled the repertoire of human leukocyte antigen class II (HLA-II)-bound peptides presented by HLA-DR diverse monocyte-derived dendritic cells pulsed with SARS-CoV-2 spike (S) protein. We identify 209 unique HLA-II-bound peptide sequences, many forming nested sets, which map to sites throughout S including glycosylated regions. Comparison of the glycosylation profile of the S protein to that of the HLA-II-bound S peptides revealed substantial trimming of glycan residues on the latter, likely introduced during antigen processing. Our data also highlight the receptor-binding motif in S1 as a HLA-DR-binding peptide-rich region. Results from this study have application in vaccine design, and will aid analysis of CD4+ T cell responses in infected individuals and vaccine recipients.
Collapse
|
14
|
Wang YT, Landeras-Bueno S, Hsieh LE, Terada Y, Kim K, Ley K, Shresta S, Saphire EO, Regla-Nava JA. Spiking Pandemic Potential: Structural and Immunological Aspects of SARS-CoV-2. Trends Microbiol 2020; 28:605-618. [PMID: 32507543 PMCID: PMC7237910 DOI: 10.1016/j.tim.2020.05.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/07/2020] [Accepted: 05/14/2020] [Indexed: 01/08/2023]
Abstract
SARS-Coronavirus-2 (SARS-CoV-2) causes Coronavirus disease 2019 (COVID-19), an infectious respiratory disease causing thousands of deaths and overwhelming public health systems. The international spread of SARS-CoV-2 is associated with the ease of global travel, and societal dynamics, immunologic naiveté of the host population, and muted innate immune responses. Based on these factors and the expanding geographic scale of the disease, the World Health Organization (WHO) declared the COVID-19 outbreak a pandemic-the first caused by a coronavirus. In this review, we summarize the current epidemiological status of COVID-19 and consider the virological and immunological lessons, animal models, and tools developed in response to prior SARS-CoV and MERS-CoV outbreaks that can serve as resources for development of SARS-CoV-2 therapeutics and vaccines. In particular, we discuss structural insights into the SARS-CoV-2 spike protein, a major determinant of transmissibility, and discuss key molecular aspects that will aid in understanding and fighting this new global threat.
Collapse
Affiliation(s)
| | | | - Li-En Hsieh
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Yutaka Terada
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Kenneth Kim
- Kord Animal Health Diagnostic Laboratory, Tennessee Department of Agriculture, Nashville, TN, USA
| | - Klaus Ley
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Sujan Shresta
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | | |
Collapse
|
15
|
Braun J, Loyal L, Frentsch M, Wendisch D, Georg P, Kurth F, Hippenstiel S, Dingeldey M, Kruse B, Fauchere F, Baysal E, Mangold M, Henze L, Lauster R, Mall MA, Beyer K, Röhmel J, Voigt S, Schmitz J, Miltenyi S, Demuth I, Müller MA, Hocke A, Witzenrath M, Suttorp N, Kern F, Reimer U, Wenschuh H, Drosten C, Corman VM, Giesecke-Thiel C, Sander LE, Thiel A. SARS-CoV-2-reactive T cells in healthy donors and patients with COVID-19. Nature 2020; 587:270-274. [PMID: 32726801 DOI: 10.1038/s41586-020-2598-9] [Citation(s) in RCA: 929] [Impact Index Per Article: 185.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused the rapidly unfolding coronavirus disease 2019 (COVID-19) pandemic1,2. Clinical manifestations of COVID-19 vary, ranging from asymptomatic infection to respiratory failure. The mechanisms that determine such variable outcomes remain unresolved. Here we investigated CD4+ T cells that are reactive against the spike glycoprotein of SARS-CoV-2 in the peripheral blood of patients with COVID-19 and SARS-CoV-2-unexposed healthy donors. We detected spike-reactive CD4+ T cells not only in 83% of patients with COVID-19 but also in 35% of healthy donors. Spike-reactive CD4+ T cells in healthy donors were primarily active against C-terminal epitopes in the spike protein, which show a higher homology to spike glycoproteins of human endemic coronaviruses, compared with N-terminal epitopes. Spike-protein-reactive T cell lines generated from SARS-CoV-2-naive healthy donors responded similarly to the C-terminal region of the spike proteins of the human endemic coronaviruses 229E and OC43, as well as that of SARS-CoV-2. This results indicate that spike-protein cross-reactive T cells are present, which were probably generated during previous encounters with endemic coronaviruses. The effect of pre-existing SARS-CoV-2 cross-reactive T cells on clinical outcomes remains to be determined in larger cohorts. However, the presence of spike-protein cross-reactive T cells in a considerable fraction of the general population may affect the dynamics of the current pandemic, and has important implications for the design and analysis of upcoming trials investigating COVID-19 vaccines.
Collapse
Affiliation(s)
- Julian Braun
- Si-M/'Der Simulierte Mensch', Technische Universität Berlin and Charité-Universitätsmedizin Berlin, Berlin, Germany.,Regenerative Immunology and Aging, BIH Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Lucie Loyal
- Si-M/'Der Simulierte Mensch', Technische Universität Berlin and Charité-Universitätsmedizin Berlin, Berlin, Germany.,Regenerative Immunology and Aging, BIH Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Marco Frentsch
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Philipp Georg
- Department of Infectious Diseases and Respiratory Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Florian Kurth
- Berlin Institute of Health (BIH), Berlin, Germany.,Department of Infectious Diseases and Respiratory Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Manuela Dingeldey
- Si-M/'Der Simulierte Mensch', Technische Universität Berlin and Charité-Universitätsmedizin Berlin, Berlin, Germany.,Regenerative Immunology and Aging, BIH Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Beate Kruse
- Si-M/'Der Simulierte Mensch', Technische Universität Berlin and Charité-Universitätsmedizin Berlin, Berlin, Germany.,Regenerative Immunology and Aging, BIH Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Florent Fauchere
- Si-M/'Der Simulierte Mensch', Technische Universität Berlin and Charité-Universitätsmedizin Berlin, Berlin, Germany.,Regenerative Immunology and Aging, BIH Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Emre Baysal
- Si-M/'Der Simulierte Mensch', Technische Universität Berlin and Charité-Universitätsmedizin Berlin, Berlin, Germany.,Regenerative Immunology and Aging, BIH Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Maike Mangold
- Si-M/'Der Simulierte Mensch', Technische Universität Berlin and Charité-Universitätsmedizin Berlin, Berlin, Germany.,Regenerative Immunology and Aging, BIH Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Larissa Henze
- Si-M/'Der Simulierte Mensch', Technische Universität Berlin and Charité-Universitätsmedizin Berlin, Berlin, Germany.,Regenerative Immunology and Aging, BIH Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Roland Lauster
- Si-M/'Der Simulierte Mensch', Technische Universität Berlin and Charité-Universitätsmedizin Berlin, Berlin, Germany.,I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcus A Mall
- Medical Biotechnology, Institute for Biotechnology, Technische Universität Berlin, Berlin, Germany.,Department of Pediatric Pulmonology, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Kirsten Beyer
- Medical Biotechnology, Institute for Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - Jobst Röhmel
- Medical Biotechnology, Institute for Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - Sebastian Voigt
- Department of Infectious Diseases, Robert Koch Institut, Berlin, Germany
| | | | | | - Ilja Demuth
- Interdisciplinary Metabolism Center, Biology of Aging (BoA) group, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Marcel A Müller
- Institute of Virology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | - Florian Kern
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, Brighton, UK.,JPT Peptide Technologies, Berlin, Germany
| | - Ulf Reimer
- JPT Peptide Technologies, Berlin, Germany
| | | | - Christian Drosten
- Department of Pediatric Pulmonology, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Institute of Virology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Victor M Corman
- Institute of Virology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | | | - Andreas Thiel
- Si-M/'Der Simulierte Mensch', Technische Universität Berlin and Charité-Universitätsmedizin Berlin, Berlin, Germany. .,Regenerative Immunology and Aging, BIH Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
16
|
Wang D, Mai J, Zhou W, Yu W, Zhan Y, Wang N, Epstein ND, Yang Y. Immunoinformatic Analysis of T- and B-Cell Epitopes for SARS-CoV-2 Vaccine Design. Vaccines (Basel) 2020; 8:vaccines8030355. [PMID: 32635180 PMCID: PMC7563688 DOI: 10.3390/vaccines8030355] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/23/2020] [Accepted: 06/30/2020] [Indexed: 12/11/2022] Open
Abstract
Currently, there is limited knowledge about the immunological profiles of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). We used computer-based immunoinformatic analysis and the newly resolved 3-dimensional (3D) structures of the SARS-CoV-2 S trimeric protein, together with analyses of the immunogenic profiles of SARS-CoV, to anticipate potential B-cell and T-cell epitopes of the SARS-CoV-2 S protein for vaccine design, particularly for peptide-driven vaccine design and serological diagnosis. Nine conserved linear B-cell epitopes and multiple discontinuous B-cell epitopes composed of 69 residues on the surface of the SARS-CoV-2 trimeric S protein were predicted to be highly antigenic. We found that the SARS-CoV-2 S protein has a different antigenic profile than that of the SARS-CoV S protein due to the variations in their primary and 3D structures. Importantly, SARS-CoV-2 may exploit an immune evasion mechanism through two point mutations in the critical and conserved linear neutralization epitope (overlap with fusion peptide) around a sparsely glycosylated area. These mutations lead to a significant decrease in the antigenicity of this epitope in the SARS-CoV-2 S protein. In addition, 62 T-cell epitopes in the SARS-CoV-2 S protein were predicted in our study. The structure-based immunoinformatic analysis for the SARS-CoV-2 S protein in this study may improve vaccine design, diagnosis, and immunotherapy against the pandemic of COVID-19.
Collapse
Affiliation(s)
- Dongliang Wang
- Hunan Provincial Key Laboratory of Protein Engineering in Animal Vaccines, Laboratory of Functional Proteomics (LFP), Research Center of Reverse Vaccinology (RCRV), College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (J.M.); (W.Z.); (W.Y.); (Y.Z.); (N.W.)
| | - Jinhui Mai
- Hunan Provincial Key Laboratory of Protein Engineering in Animal Vaccines, Laboratory of Functional Proteomics (LFP), Research Center of Reverse Vaccinology (RCRV), College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (J.M.); (W.Z.); (W.Y.); (Y.Z.); (N.W.)
| | - Wenfeng Zhou
- Hunan Provincial Key Laboratory of Protein Engineering in Animal Vaccines, Laboratory of Functional Proteomics (LFP), Research Center of Reverse Vaccinology (RCRV), College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (J.M.); (W.Z.); (W.Y.); (Y.Z.); (N.W.)
| | - Wanting Yu
- Hunan Provincial Key Laboratory of Protein Engineering in Animal Vaccines, Laboratory of Functional Proteomics (LFP), Research Center of Reverse Vaccinology (RCRV), College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (J.M.); (W.Z.); (W.Y.); (Y.Z.); (N.W.)
- State Key Laboratory of Membrane Biology, Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
| | - Yang Zhan
- Hunan Provincial Key Laboratory of Protein Engineering in Animal Vaccines, Laboratory of Functional Proteomics (LFP), Research Center of Reverse Vaccinology (RCRV), College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (J.M.); (W.Z.); (W.Y.); (Y.Z.); (N.W.)
| | - Naidong Wang
- Hunan Provincial Key Laboratory of Protein Engineering in Animal Vaccines, Laboratory of Functional Proteomics (LFP), Research Center of Reverse Vaccinology (RCRV), College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (J.M.); (W.Z.); (W.Y.); (Y.Z.); (N.W.)
| | - Neal D. Epstein
- Cell and Developmental Biology Center, NHLBI, NIH, Bethesda, MD 20892, USA
- Correspondence: (N.D.E.); (Y.Y.)
| | - Yi Yang
- Hunan Provincial Key Laboratory of Protein Engineering in Animal Vaccines, Laboratory of Functional Proteomics (LFP), Research Center of Reverse Vaccinology (RCRV), College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (D.W.); (J.M.); (W.Z.); (W.Y.); (Y.Z.); (N.W.)
- Correspondence: (N.D.E.); (Y.Y.)
| |
Collapse
|
17
|
Smith CC, Entwistle S, Willis C, Vensko S, Beck W, Garness J, Sambade M, Routh E, Olsen K, Kodysh J, O’Donnell T, Haber C, Heiss K, Stadler V, Garrison E, Grant OC, Woods RJ, Heise M, Vincent BG, Rubinsteyn A. Landscape and Selection of Vaccine Epitopes in SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.06.04.135004. [PMID: 32577654 PMCID: PMC7302209 DOI: 10.1101/2020.06.04.135004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
There is an urgent need for a vaccine with efficacy against SARS-CoV-2. We hypothesize that peptide vaccines containing epitope regions optimized for concurrent B cell, CD4+ T cell, and CD8+ T cell stimulation would drive both humoral and cellular immunity with high specificity, potentially avoiding undesired effects such as antibody-dependent enhancement (ADE). Additionally, such vaccines can be rapidly manufactured in a distributed manner. In this study, we combine computational prediction of T cell epitopes, recently published B cell epitope mapping studies, and epitope accessibility to select candidate peptide vaccines for SARS-CoV-2. We begin with an exploration of the space of possible T cell epitopes in SARS-CoV-2 with interrogation of predicted HLA-I and HLA-II ligands, overlap between predicted ligands, protein source, as well as concurrent human/murine coverage. Beyond MHC affinity, T cell vaccine candidates were further refined by predicted immunogenicity, viral source protein abundance, sequence conservation, coverage of high frequency HLA alleles and co-localization of CD4+ and CD8+ T cell epitopes. B cell epitope regions were chosen from linear epitope mapping studies of convalescent patient serum, followed by filtering to select regions with surface accessibility, high sequence conservation, spatial localization near functional domains of the spike glycoprotein, and avoidance of glycosylation sites. From 58 initial candidates, three B cell epitope regions were identified. By combining these B cell and T cell analyses, as well as a manufacturability heuristic, we propose a set of SARS-CoV-2 vaccine peptides for use in subsequent murine studies and clinical trials.
Collapse
Affiliation(s)
- Christof C. Smith
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Sarah Entwistle
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Caryn Willis
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Steven Vensko
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Wolfgang Beck
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jason Garness
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Maria Sambade
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Eric Routh
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kelly Olsen
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Julia Kodysh
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Timothy O’Donnell
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | | | - Erik Garrison
- Genomics Institute, University of California, Santa Cruz, California
| | - Oliver C. Grant
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia
| | - Robert J. Woods
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia
| | - Mark Heise
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Genetics, UNC School of Medicine, Chapel Hill, North Carolina
| | - Benjamin G. Vincent
- Department of Microbiology and Immunology, UNC School of Medicine, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Computational Medicine Program, UNC School of Medicine, Chapel Hill, North Carolina
- Curriculum in Bioinformatics and Computational Biology, UNC School of Medicine, Chapel Hill, North Carolina
- Division of Hematology/Oncology, Department of Medicine, UNC School of Medicine, Chapel Hill, North Carolina
| | - Alex Rubinsteyn
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Genetics, UNC School of Medicine, Chapel Hill, North Carolina
- Computational Medicine Program, UNC School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
18
|
Shivarov V, Petrov PK, Pashov AD. Potential SARS-CoV-2 Preimmune IgM Epitopes. Front Immunol 2020; 11:932. [PMID: 32425955 PMCID: PMC7203467 DOI: 10.3389/fimmu.2020.00932] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 04/21/2020] [Indexed: 11/13/2022] Open
Abstract
While studying the human public IgM igome as represented by a library of 224,087 linear mimotopes, three exact matches to peptides in the proteins of SARS-CoV-2 were found: two in the open reading frame 1ab and one in the spike protein. Joining the efforts to fast track SARS-CoV-2 vaccine development, here we describe briefly these potential epitopes in comparison to mimotopes representing peptides of SARS-CoV, HCoV 229E and OC43.
Collapse
Affiliation(s)
- Velizar Shivarov
- Laboratory of Clinical Immunology and Department of Clinical Hematology, Sofiamed University Hospital, Sofia, Bulgaria
- Faculty of Biology, Sofia University, Sofia, Bulgaria
| | - Peter K. Petrov
- Department of Analysis, Geometry and Topology, Institute of Mathematics and Informatics, Bulgarian Academy of Sciences (BAS), Sofia, Bulgaria
| | - Anastas D. Pashov
- Department of Immunology, Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences (BAS), Sofia, Bulgaria
| |
Collapse
|
19
|
Grifoni A, Sidney J, Zhang Y, Scheuermann RH, Peters B, Sette A. A Sequence Homology and Bioinformatic Approach Can Predict Candidate Targets for Immune Responses to SARS-CoV-2. Cell Host Microbe 2020; 27:671-680.e2. [PMID: 32183941 PMCID: PMC7142693 DOI: 10.1016/j.chom.2020.03.002] [Citation(s) in RCA: 734] [Impact Index Per Article: 146.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 02/26/2020] [Accepted: 03/05/2020] [Indexed: 10/26/2022]
Abstract
Effective countermeasures against the recent emergence and rapid expansion of the 2019 novel coronavirus (SARS-CoV-2) require the development of data and tools to understand and monitor its spread and immune responses to it. However, little information is available about the targets of immune responses to SARS-CoV-2. We used the Immune Epitope Database and Analysis Resource (IEDB) to catalog available data related to other coronaviruses. This includes SARS-CoV, which has high sequence similarity to SARS-CoV-2 and is the best-characterized coronavirus in terms of epitope responses. We identified multiple specific regions in SARS-CoV-2 that have high homology to the SARS-CoV virus. Parallel bioinformatic predictions identified a priori potential B and T cell epitopes for SARS-CoV-2. The independent identification of the same regions using two approaches reflects the high probability that these regions are promising targets for immune recognition of SARS-CoV-2. These predictions can facilitate effective vaccine design against this virus of high priority.
Collapse
Affiliation(s)
- Alba Grifoni
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - John Sidney
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Yun Zhang
- J. Craig Venter Institute, La Jolla, CA 92037, USA
| | - Richard H Scheuermann
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; J. Craig Venter Institute, La Jolla, CA 92037, USA; Department of Pathology, University of California, San Diego, San Diego, CA 92093, USA
| | - Bjoern Peters
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA.
| |
Collapse
|
20
|
Cabrera-Perez J, Badovinac VP, Griffith TS. Enteric immunity, the gut microbiome, and sepsis: Rethinking the germ theory of disease. Exp Biol Med (Maywood) 2016; 242:127-139. [PMID: 27633573 DOI: 10.1177/1535370216669610] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Sepsis is a poorly understood syndrome of systemic inflammation responsible for hundreds of thousands of deaths every year. The integrity of the gut epithelium and competence of adaptive immune responses are notoriously compromised during sepsis, and the prevalent assumption in the scientific and medical community is that intestinal commensals have a detrimental role in the systemic inflammation and susceptibility to nosocomial infections seen in critically ill, septic patients. However, breakthroughs in the last decade provide strong credence to the idea that our mucosal microbiome plays an essential role in adaptive immunity, where a human host and its prokaryotic colonists seem to exist in a carefully negotiated armistice with compromises and benefits that go both ways. In this review, we re-examine the notion that intestinal contents are the driving force of critical illness. An overview of the interaction between the microbiome and the immune system is provided, with a special focus on the impact of commensals in priming and the careful balance between normal intestinal flora and pathogenic organisms residing in the gut microbiome. Based on the data in hand, we hypothesize that sepsis induces imbalances in microbial populations residing in the gut, along with compromises in epithelial integrity. As a result, normal antigen sampling becomes impaired, and proliferative cues are intermixed with inhibitory signals. This situates the microbiome, the gut, and its complex immune network of cells and bacteria, at the center of aberrant immune responses during and after sepsis.
Collapse
Affiliation(s)
- Javier Cabrera-Perez
- 1 Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota Medical School, Minneapolis, MN 55455, USA.,2 Medical Scientist Training Program, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Vladimir P Badovinac
- 3 Department of Pathology, University of Minnesota Medical School, Minneapolis, MN 55455, USA.,4 Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Thomas S Griffith
- 1 Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota Medical School, Minneapolis, MN 55455, USA.,5 Department of Urology, University of Minnesota Medical School, Minneapolis, MN 55455, USA.,6 Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA.,7 Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, MN 55455, USA.,8 Minneapolis VA Health Care System, Minneapolis, MN 55417, USA
| |
Collapse
|
21
|
Huang J, Cao Y, Bu X, Wu C. Residue analysis of a CTL epitope of SARS-CoV spike protein by IFN-gamma production and bioinformatics prediction. BMC Immunol 2012; 13:50. [PMID: 22963340 PMCID: PMC3575293 DOI: 10.1186/1471-2172-13-50] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 08/31/2012] [Indexed: 01/15/2023] Open
Abstract
Background Severe acute respiratory syndrome (SARS) is an emerging infectious disease caused by the novel coronavirus SARS-CoV. The T cell epitopes of the SARS CoV spike protein are well known, but no systematic evaluation of the functional and structural roles of each residue has been reported for these antigenic epitopes. Analysis of the functional importance of side-chains by mutational study may exaggerate the effect by imposing a structural disturbance or an unusual steric, electrostatic or hydrophobic interaction. Results We demonstrated that N50 could induce significant IFN-gamma response from SARS-CoV S DNA immunized mice splenocytes by the means of ELISA, ELISPOT and FACS. Moreover, S366-374 was predicted to be an optimal epitope by bioinformatics tools: ANN, SMM, ARB and BIMAS, and confirmed by IFN-gamma response induced by a series of S358-374-derived peptides. Furthermore, each of S366-374 was replaced by alanine (A), lysine (K) or aspartic acid (D), respectively. ANN was used to estimate the binding affinity of single S366-374 mutants to H-2 Kd. Y367 and L374 were predicated to possess the most important role in peptide binding. Additionally, these one residue mutated peptides were synthesized, and IFN-gamma production induced by G368, V369, A371, T372 and K373 mutated S366-374 were decreased obviously. Conclusions We demonstrated that S366-374 is an optimal H-2 Kd CTL epitope in the SARS CoV S protein. Moreover, Y367, S370, and L374 are anchors in the epitope, while C366, G368, V369, A371, T372, and K373 may directly interact with TCR on the surface of CD8-T cells.
Collapse
Affiliation(s)
- Jun Huang
- Institute of Immunology, Zhongshan School of Medicine, Key Laboratory of Tropical Disease Control Research of Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | | | | | | |
Collapse
|
22
|
Abstract
MHC class II tetramers have emerged as an important tool for characterization of the specificity and phenotype of CD4 T cell immune responses, useful in a large variety of disease and vaccine studies. Issues of specific T cell frequency, biodistribution, and avidity, coupled with the large genetic diversity of potential class II restriction elements, require targeted experimental design. Translational opportunities for immune disease monitoring are driving the rapid development of HLA class II tetramer use in clinical applications, together with innovations in tetramer production and epitope discovery.
Collapse
|
23
|
Shen C, He Y, Meng F, Cheng K, Zhang D, Miao F, Zhang J. Characterization of MHC/peptide complexes refolded by a one-step ion-exchange chromatography. J Immunol Methods 2011; 369:81-90. [DOI: 10.1016/j.jim.2011.04.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Revised: 02/14/2011] [Accepted: 04/21/2011] [Indexed: 01/12/2023]
|
24
|
Keicho N, Itoyama S, Kashiwase K, Phi NC, Long HT, Ha LD, Ban VV, Hoa BK, Hang NTL, Hijikata M, Sakurada S, Satake M, Tokunaga K, Sasazuki T, Quy T. Association of human leukocyte antigen class II alleles with severe acute respiratory syndrome in the Vietnamese population. Hum Immunol 2009; 70:527-31. [PMID: 19445991 PMCID: PMC7132661 DOI: 10.1016/j.humimm.2009.05.006] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2009] [Revised: 05/07/2009] [Accepted: 05/08/2009] [Indexed: 02/07/2023]
Abstract
Excessive immune response is believed to play a role in the development of severe acute respiratory syndrome (SARS). Inhomogeneous spread of SARS led one to think of an Asian genetic predisposition and contribution of human leukocyte antigen (HLA) to the disease susceptibility. However, past case-control studies showed inconsistent results. In Viet Nam, of 62 patients with SARS, 44 participated in the present study together with 103 individuals who had contact with SARS patients and 50 without contact history. HLA-DRB1*12 was more frequently shown in SARS patients than in controls (corrected p = 0.042). HLA-DRB1*1202, the predominant allele in the Vietnamese population showed the strongest association with SARS in a dominant model (corrected p = 0.0065 and 0.0052, depending on the controls to be compared). Our results and accumulated data on HLA in the Asian populations would help in the understanding of associations with emerging infectious diseases.
Collapse
Affiliation(s)
- Naoto Keicho
- Department of Respiratory Diseases, Research Institute, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|