1
|
Kupke A, Volz A, Dietzel E, Freudenstein A, Schmidt J, Shams-Eldin H, Jany S, Sauerhering L, Krähling V, Gellhorn Serra M, Herden C, Eickmann M, Becker S, Sutter G. Protective CD8+ T Cell Response Induced by Modified Vaccinia Virus Ankara Delivering Ebola Virus Nucleoprotein. Vaccines (Basel) 2022; 10:vaccines10040533. [PMID: 35455282 PMCID: PMC9027530 DOI: 10.3390/vaccines10040533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/23/2022] [Accepted: 03/25/2022] [Indexed: 02/01/2023] Open
Abstract
The urgent need for vaccines against Ebola virus (EBOV) was underscored by the large outbreak in West Africa (2014–2016). Since then, several promising vaccine candidates have been tested in pre-clinical and clinical studies. As a result, two vaccines were approved for human use in 2019/2020, of which one includes a heterologous adenovirus/Modified Vaccinia virus Ankara (MVA) prime-boost regimen. Here, we tested new vaccine candidates based on the recombinant MVA vector, encoding the EBOV nucleoprotein (MVA-EBOV-NP) or glycoprotein (MVA-EBOV-GP) for their efficacy after homologous prime-boost immunization in mice. Our aim was to investigate the role of each antigen in terms of efficacy and correlates of protection. Sera of mice vaccinated with MVA-EBOV-GP were virus-neutralizing and MVA-EBOV-NP immunization readily elicited interferon-γ-producing NP-specific CD8+ T cells. While mock-vaccinated mice succumbed to EBOV infection, all vaccinated mice survived and showed drastically decreased viral loads in sera and organs. In addition, MVA-EBOV-NP vaccinated mice became susceptible to lethal EBOV infection after depletion of CD8+ T cells prior to challenge. This study highlights the potential of MVA-based vaccines to elicit humoral immune responses as well as a strong and protective CD8+ T cell response and contributes to understanding the possible underlying mechanisms.
Collapse
Affiliation(s)
- Alexandra Kupke
- Institute of Virology, Philipps University Marburg, 35043 Marburg, Germany; (A.K.); (E.D.); (J.S.); (H.S.-E.); (L.S.); (V.K.); (M.G.S.); (M.E.)
- German Center for Infection Research, Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Asisa Volz
- Institute of Virology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany;
- German Center for Infection Research, Partner Site Munich, 80539 Munich, Germany;
| | - Erik Dietzel
- Institute of Virology, Philipps University Marburg, 35043 Marburg, Germany; (A.K.); (E.D.); (J.S.); (H.S.-E.); (L.S.); (V.K.); (M.G.S.); (M.E.)
- German Center for Infection Research, Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Astrid Freudenstein
- Division of Virology, Institute for Infectious Diseases and Zoonoses, LMU Munich, 80539 Munich, Germany; (A.F.); (S.J.)
| | - Jörg Schmidt
- Institute of Virology, Philipps University Marburg, 35043 Marburg, Germany; (A.K.); (E.D.); (J.S.); (H.S.-E.); (L.S.); (V.K.); (M.G.S.); (M.E.)
- German Center for Infection Research, Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Hosam Shams-Eldin
- Institute of Virology, Philipps University Marburg, 35043 Marburg, Germany; (A.K.); (E.D.); (J.S.); (H.S.-E.); (L.S.); (V.K.); (M.G.S.); (M.E.)
| | - Sylvia Jany
- Division of Virology, Institute for Infectious Diseases and Zoonoses, LMU Munich, 80539 Munich, Germany; (A.F.); (S.J.)
| | - Lucie Sauerhering
- Institute of Virology, Philipps University Marburg, 35043 Marburg, Germany; (A.K.); (E.D.); (J.S.); (H.S.-E.); (L.S.); (V.K.); (M.G.S.); (M.E.)
- German Center for Infection Research, Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Verena Krähling
- Institute of Virology, Philipps University Marburg, 35043 Marburg, Germany; (A.K.); (E.D.); (J.S.); (H.S.-E.); (L.S.); (V.K.); (M.G.S.); (M.E.)
- German Center for Infection Research, Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Michelle Gellhorn Serra
- Institute of Virology, Philipps University Marburg, 35043 Marburg, Germany; (A.K.); (E.D.); (J.S.); (H.S.-E.); (L.S.); (V.K.); (M.G.S.); (M.E.)
| | - Christiane Herden
- Institute of Veterinary Pathology, Justus Liebig University Giessen, 35392 Giessen, Germany;
| | - Markus Eickmann
- Institute of Virology, Philipps University Marburg, 35043 Marburg, Germany; (A.K.); (E.D.); (J.S.); (H.S.-E.); (L.S.); (V.K.); (M.G.S.); (M.E.)
- German Center for Infection Research, Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Stephan Becker
- Institute of Virology, Philipps University Marburg, 35043 Marburg, Germany; (A.K.); (E.D.); (J.S.); (H.S.-E.); (L.S.); (V.K.); (M.G.S.); (M.E.)
- German Center for Infection Research, Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
- Correspondence:
| | - Gerd Sutter
- German Center for Infection Research, Partner Site Munich, 80539 Munich, Germany;
- Division of Virology, Institute for Infectious Diseases and Zoonoses, LMU Munich, 80539 Munich, Germany; (A.F.); (S.J.)
| |
Collapse
|
2
|
Structural and Functional Aspects of Ebola Virus Proteins. Pathogens 2021; 10:pathogens10101330. [PMID: 34684279 PMCID: PMC8538763 DOI: 10.3390/pathogens10101330] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/11/2021] [Accepted: 10/14/2021] [Indexed: 01/14/2023] Open
Abstract
Ebola virus (EBOV), member of genus Ebolavirus, family Filoviridae, have a non-segmented, single-stranded RNA that contains seven genes: (a) nucleoprotein (NP), (b) viral protein 35 (VP35), (c) VP40, (d) glycoprotein (GP), (e) VP30, (f) VP24, and (g) RNA polymerase (L). All genes encode for one protein each except GP, producing three pre-proteins due to the transcriptional editing. These pre-proteins are translated into four products, namely: (a) soluble secreted glycoprotein (sGP), (b) Δ-peptide, (c) full-length transmembrane spike glycoprotein (GP), and (d) soluble small secreted glycoprotein (ssGP). Further, shed GP is released from infected cells due to cleavage of GP by tumor necrosis factor α-converting enzyme (TACE). This review presents a detailed discussion on various functional aspects of all EBOV proteins and their residues. An introduction to ebolaviruses and their life cycle is also provided for clarity of the available analysis. We believe that this review will help understand the roles played by different EBOV proteins in the pathogenesis of the disease. It will help in targeting significant protein residues for therapeutic and multi-protein/peptide vaccine development.
Collapse
|
3
|
Powlson J, Wright D, Zeltina A, Giza M, Nielsen M, Rampling T, Venkatrakaman N, Bowden TA, Hill AVS, Ewer KJ. Characterization of Antigenic MHC-Class-I-Restricted T Cell Epitopes in the Glycoprotein of Ebolavirus. Cell Rep 2020; 29:2537-2545.e3. [PMID: 31775024 PMCID: PMC6899439 DOI: 10.1016/j.celrep.2019.10.105] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/20/2019] [Accepted: 10/25/2019] [Indexed: 11/05/2022] Open
Abstract
Ebolavirus causes highly lethal hemorrhagic fever in humans. The envelope-displayed viral glycoprotein (GP) is the primary target of humoral immunity induced by natural exposure and vaccination. No T cell epitopes in the GP have been characterized in humans. A phase I clinical trial of a heterologous prime-boost vaccination regime with viral vectors encoding filovirus antigens elicits humoral and T cell responses in vaccinees. The most frequently recognized peptide pools are deconvoluted to identify the minimal epitopes recognized by antigen-specific T cells. We characterize nine immunogenic epitopes on the Ebolavirus GP. Histocompatibility leukocyte antigen (HLA) typing with in silico epitope analysis determines the likely MHC class I restriction elements. Thirteen HLA-A and -B alleles are predicted to present the identified CD8+ T cell epitopes, suggesting promiscuous recognition and a broad immune response. Delivery of the Ebolavirus GP antigen by using a heterologous prime-boost approach is immunogenic in genetically diverse human populations, with responses against multiple epitopes. Vaccination induces high T cell responses to the Ebola virus glycoprotein in humans Eight CD8+ epitopes were defined, recognized through multiple MHC class I alleles Responses match those observed in Ebola survivors and could boost vaccine efficacy
Collapse
Affiliation(s)
- Jonathan Powlson
- The Jenner Institute, Old Road Campus Research Building, University of Oxford, Oxford OX3 7DQ, UK
| | - Daniel Wright
- The Jenner Institute, Old Road Campus Research Building, University of Oxford, Oxford OX3 7DQ, UK
| | - Antra Zeltina
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Mark Giza
- The Jenner Institute, Old Road Campus Research Building, University of Oxford, Oxford OX3 7DQ, UK
| | - Morten Nielsen
- Department of Health Technology, The Technical University of Denmark, Anker Engelunds Vej 1 Bygning 101A, 2800 Kgs Lyngby, Denmark
| | - Tommy Rampling
- The Jenner Institute, Old Road Campus Research Building, University of Oxford, Oxford OX3 7DQ, UK
| | - Navin Venkatrakaman
- The Jenner Institute, Old Road Campus Research Building, University of Oxford, Oxford OX3 7DQ, UK
| | - Thomas A Bowden
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Adrian V S Hill
- The Jenner Institute, Old Road Campus Research Building, University of Oxford, Oxford OX3 7DQ, UK
| | - Katie J Ewer
- The Jenner Institute, Old Road Campus Research Building, University of Oxford, Oxford OX3 7DQ, UK.
| |
Collapse
|
4
|
Jain S, Baranwal M. Conserved peptide vaccine candidates containing multiple Ebola nucleoprotein epitopes display interactions with diverse HLA molecules. Med Microbiol Immunol 2019; 208:227-238. [PMID: 30790057 DOI: 10.1007/s00430-019-00584-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 02/11/2019] [Indexed: 10/27/2022]
Abstract
Immunoinformatics has come by leaps and bounds to finding potent vaccine candidates against various pathogens. In the current study, a combination of different T (CD4+ and CD8+) and B cell epitope prediction tools was applied to find peptides containing multiple epitopes against Ebola nucleoprotein (NP) and the presentation of peptides to human leukocyte antigen (HLA) molecules was analyzed by prediction, docking and population coverage tools. Further, potential peptides were analyzed by ELISA for peptide induced IFN-γ secretion in peripheral blood mononuclear cells isolated from healthy volunteers. Six peptides were obtained after merging the overlapping multiple HLA I (CD8+) and II (CD4+) restricted T cell epitopes as well as B cell epitopes and eliminating the peptides liable to generate autoimmune and allergic response. All peptides displayed 100% conservancy in Zaire ebolavirus. In other Ebola virus species (Sudan, Bundibugyo and Taï forest) and Filoviridae members (Lloviuvirus and Margburgvirus), some peptides were found to be conserved with minor variations. Prediction tools confirmed the ability of predicted peptides to bind with diverse HLA (HLA-A, HLA-B, HLA-DP, HLA-DQ and HLA-DR) alleles. CABS-dock results displayed that the average root mean square deviation (RMSD) value was less than three in majority of cases representing strong binding affinity with HLA alleles. Population coverage analysis predicted high coverage (> 85%) for expected immune response in four continents (Africa, America, Asia and Europe). Nine out of ten blood samples exhibited enhanced IFN-γ secretion for two peptides (P2 and P3). Thus, the identified NP peptides can be considered as potential synthetic vaccine candidates against Ebola virus.
Collapse
Affiliation(s)
- Sahil Jain
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India.
| |
Collapse
|
5
|
Perdomo-Celis F, Salvato MS, Medina-Moreno S, Zapata JC. T-Cell Response to Viral Hemorrhagic Fevers. Vaccines (Basel) 2019; 7:E11. [PMID: 30678246 PMCID: PMC6466054 DOI: 10.3390/vaccines7010011] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/15/2019] [Accepted: 01/19/2019] [Indexed: 12/22/2022] Open
Abstract
Viral hemorrhagic fevers (VHF) are a group of clinically similar diseases that can be caused by enveloped RNA viruses primarily from the families Arenaviridae, Filoviridae, Hantaviridae, and Flaviviridae. Clinically, this group of diseases has in common fever, fatigue, dizziness, muscle aches, and other associated symptoms that can progress to vascular leakage, bleeding and multi-organ failure. Most of these viruses are zoonotic causing asymptomatic infections in the primary host, but in human beings, the infection can be lethal. Clinical and experimental evidence suggest that the T-cell response is needed for protection against VHF, but can also cause damage to the host, and play an important role in disease pathogenesis. Here, we present a review of the T-cell immune responses to VHF and insights into the possible ways to improve counter-measures for these viral agents.
Collapse
Affiliation(s)
- Federico Perdomo-Celis
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, 050010, Colombia.
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA.
| | - Maria S Salvato
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA.
| | - Sandra Medina-Moreno
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA.
| | - Juan C Zapata
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA.
| |
Collapse
|
6
|
Identification of novel HLA-A11-restricted T-cell epitopes in the Ebola virus nucleoprotein. Microbes Infect 2018; 21:56-62. [PMID: 29775667 DOI: 10.1016/j.micinf.2018.04.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/20/2018] [Accepted: 04/26/2018] [Indexed: 12/17/2022]
Abstract
The Ebola virus (EBOV) is a very contagious virus that is highly fatal in humans and animals. The largest epidemic was in West Africa in 2014, in which over 11,000 people died. However, to date, there are no licensed vaccines against it. Studies show that CD4+ and CD8+ T-cell responses, especially cytotoxic T-lymphocyte (CTL) responses, play key roles in protecting individuals from EBOV infection. Since HLA-restricted epitope vaccines are likely to be effective and safe immunization strategies for infectious diseases, the present study screened for CTL epitopes in the EBOV-nucleoprotein that are restricted by HLA-A11 (a common allele in Chinese people). Predictive computer analysis of the amino-acid sequence of EBOV-nucleoprotein identified ten putative HLA-A11-restricted epitopes. ELISPOT assay of immunized HLA-A11/DR1 transgenic mice showed that five (GR-9, VR-9, EK-9, PK-9, and RK-9) induced effective CTL responses. Additional epitope analyses will aid the design of epitope vaccines against EBOV.
Collapse
|
7
|
Lehrer AT, Wong TAS, Lieberman MM, Humphreys T, Clements DE, Bakken RR, Hart MK, Pratt WD, Dye JM. Recombinant proteins of Zaire ebolavirus induce potent humoral and cellular immune responses and protect against live virus infection in mice. Vaccine 2018; 36:3090-3100. [PMID: 28216187 PMCID: PMC8426131 DOI: 10.1016/j.vaccine.2017.01.068] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/17/2017] [Accepted: 01/30/2017] [Indexed: 01/26/2023]
Abstract
Infections with filoviruses in humans are highly virulent, causing hemorrhagic fevers which result in up to 90% mortality. In addition to natural infections, the ability to use these viruses as bioterrorist weapons is of significant concern. Currently, there are no licensed vaccines or therapeutics available to combat these infections. The pathogenesis of disease involves the dysregulation of the host's immune system, which results in impairment of the innate and adaptive immune responses, with subsequent development of lymphopenia, thrombocytopenia, hemorrhage, and death. Questions remain with regard to the few survivors of infection, who manage to mount an effective adaptive immune response. These questions concern the humoral and cellular components of this response, and whether such a response can be elicited by an appropriate prophylactic vaccine. The data reported herein describe the production and evaluation of a recombinant subunit Ebola virus vaccine candidate consisting of insect cell expressed Zaire ebolavirus (EBOV) surface glycoprotein (GP) and the matrix proteins VP24 and VP40. The recombinant subunit proteins are shown to be highly immunogenic in mice, yielding both humoral and cellular responses, as well as highly efficacious, providing up to 100% protection against a lethal challenge with live virus. These results demonstrate proof of concept for such a recombinant non-replicating vaccine candidate in the mouse model of EBOV which helps to elucidate immune correlates of protection and warrants further development.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- Ebola Vaccines/immunology
- Ebolavirus
- Female
- Hemorrhagic Fever, Ebola/immunology
- Hemorrhagic Fever, Ebola/prevention & control
- Immunity, Cellular
- Immunity, Humoral
- Mice
- Mice, Inbred BALB C
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/immunology
- Viral Envelope Proteins/immunology
- Viral Matrix Proteins/immunology
- Viral Proteins/immunology
Collapse
Affiliation(s)
- Axel T Lehrer
- PanThera Biopharma, LLC, Aiea, HI 96701, United States.
| | | | | | | | | | - Russell R Bakken
- US Army Medical Research Institute for Infectious Diseases, Fort Detrick, MD 21702, United States
| | - Mary Kate Hart
- US Army Medical Research Institute for Infectious Diseases, Fort Detrick, MD 21702, United States
| | - William D Pratt
- US Army Medical Research Institute for Infectious Diseases, Fort Detrick, MD 21702, United States
| | - John M Dye
- US Army Medical Research Institute for Infectious Diseases, Fort Detrick, MD 21702, United States
| |
Collapse
|
8
|
Anticoli S, Manfredi F, Chiozzini C, Arenaccio C, Olivetta E, Ferrantelli F, Capocefalo A, Falcone E, Ruggieri A, Federico M. An Exosome-Based Vaccine Platform Imparts Cytotoxic T Lymphocyte Immunity Against Viral Antigens. Biotechnol J 2018; 13:e1700443. [DOI: 10.1002/biot.201700443] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 11/17/2017] [Indexed: 01/01/2023]
Affiliation(s)
- Simona Anticoli
- National Center for Global Health Istituto Superiore di Sanità; Viale Regina Elena, 299 00161, Rome Italy
| | - Francesco Manfredi
- National Center for Global Health Istituto Superiore di Sanità; Viale Regina Elena, 299 00161, Rome Italy
| | - Chiara Chiozzini
- National Center for Global Health Istituto Superiore di Sanità; Viale Regina Elena, 299 00161, Rome Italy
| | - Claudia Arenaccio
- National Center for Global Health Istituto Superiore di Sanità; Viale Regina Elena, 299 00161, Rome Italy
| | - Eleonora Olivetta
- National Center for Global Health Istituto Superiore di Sanità; Viale Regina Elena, 299 00161, Rome Italy
| | - Flavia Ferrantelli
- National Center for Global Health Istituto Superiore di Sanità; Viale Regina Elena, 299 00161, Rome Italy
| | - Antonio Capocefalo
- Department of Veterinary Public Health & Food Safety Istituto Superiore di Sanità; Viale Regina Elena, 299 00161, Rome Italy
| | - Emiliana Falcone
- Department of Veterinary Public Health & Food Safety Istituto Superiore di Sanità; Viale Regina Elena, 299 00161, Rome Italy
| | - Anna Ruggieri
- Department of Veterinary Public Health & Food Safety Istituto Superiore di Sanità; Viale Regina Elena, 299 00161, Rome Italy
| | - Maurizio Federico
- National Center for Global Health Istituto Superiore di Sanità; Viale Regina Elena, 299 00161, Rome Italy
| |
Collapse
|
9
|
Ruibal P, Oestereich L, Lüdtke A, Becker-Ziaja B, Wozniak DM, Kerber R, Korva M, Cabeza-Cabrerizo M, Bore JA, Koundouno FR, Duraffour S, Weller R, Thorenz A, Cimini E, Viola D, Agrati C, Repits J, Afrough B, Cowley LA, Ngabo D, Hinzmann J, Mertens M, Vitoriano I, Logue CH, Boettcher JP, Pallasch E, Sachse A, Bah A, Nitzsche K, Kuisma E, Michel J, Holm T, Zekeng EG, García-Dorival I, Wölfel R, Stoecker K, Fleischmann E, Strecker T, Di Caro A, Avšič-Županc T, Kurth A, Meschi S, Mély S, Newman E, Bocquin A, Kis Z, Kelterbaum A, Molkenthin P, Carletti F, Portmann J, Wolff S, Castilletti C, Schudt G, Fizet A, Ottowell LJ, Herker E, Jacobs T, Kretschmer B, Severi E, Ouedraogo N, Lago M, Negredo A, Franco L, Anda P, Schmiedel S, Kreuels B, Wichmann D, Addo MM, Lohse AW, De Clerck H, Nanclares C, Jonckheere S, Van Herp M, Sprecher A, Xiaojiang G, Carrington M, Miranda O, Castro CM, Gabriel M, Drury P, Formenty P, Diallo B, Koivogui L, Magassouba N, Carroll MW, Günther S, Muñoz-Fontela C. Unique human immune signature of Ebola virus disease in Guinea. Nature 2016; 533:100-4. [PMID: 27147028 PMCID: PMC4876960 DOI: 10.1038/nature17949] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 04/01/2016] [Indexed: 01/20/2023]
Abstract
Despite the magnitude of the Ebola virus disease (EVD) outbreak in West Africa, there is still a fundamental lack of knowledge about the pathophysiology of EVD. In particular, very little is known about human immune responses to Ebola virus. Here we evaluate the physiology of the human T cell immune response in EVD patients at the time of admission to the Ebola Treatment Center in Guinea, and longitudinally until discharge or death. Through the use of multiparametric flow cytometry established by the European Mobile Laboratory in the field, we identify an immune signature that is unique in EVD fatalities. Fatal EVD was characterized by a high percentage of CD4(+) and CD8(+) T cells expressing the inhibitory molecules CTLA-4 and PD-1, which correlated with elevated inflammatory markers and high virus load. Conversely, surviving individuals showed significantly lower expression of CTLA-4 and PD-1 as well as lower inflammation, despite comparable overall T cell activation. Concomitant with virus clearance, survivors mounted a robust Ebola-virus-specific T cell response. Our findings suggest that dysregulation of the T cell response is a key component of EVD pathophysiology.
Collapse
Affiliation(s)
- Paula Ruibal
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, 20251 Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, 20359 Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, and Marburg, Germany
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
| | - Lisa Oestereich
- Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, 20359 Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, and Marburg, Germany
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
| | - Anja Lüdtke
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, 20251 Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, 20359 Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, and Marburg, Germany
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
| | - Beate Becker-Ziaja
- Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, 20359 Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, and Marburg, Germany
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
| | - David M Wozniak
- Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, 20359 Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, and Marburg, Germany
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
| | - Romy Kerber
- Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, 20359 Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, and Marburg, Germany
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
| | - Miša Korva
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Mar Cabeza-Cabrerizo
- Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, 20359 Hamburg, Germany
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
| | - Joseph A Bore
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
| | - Fara Raymond Koundouno
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
| | - Sophie Duraffour
- Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, 20359 Hamburg, Germany
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
| | - Romy Weller
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Institute of Experimental Virology, Twincore, Center for Experimental and Clinical Infection Research, 30625 Hannover, Germany
| | - Anja Thorenz
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Hannover Medical School, 30625 Hannover, Germany
| | - Eleonora Cimini
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- National Institute for Infectious Diseases 'Lazzaro Spallanzani', 00149 Rome, Italy
| | - Domenico Viola
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- National Institute for Infectious Diseases 'Lazzaro Spallanzani', 00149 Rome, Italy
| | - Chiara Agrati
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- National Institute for Infectious Diseases 'Lazzaro Spallanzani', 00149 Rome, Italy
| | - Johanna Repits
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
| | - Babak Afrough
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Public Health England, Porton Down, Salisbury SP4 0JG, UK
| | - Lauren A Cowley
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Public Health England, Colindale Ave, London NW9 5EQ, UK
| | - Didier Ngabo
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Public Health England, Porton Down, Salisbury SP4 0JG, UK
| | - Julia Hinzmann
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Robert Koch Institute, 13353 Berlin, Germany
| | - Marc Mertens
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Friedrich Loeffler Institute, 17493 Greifswald-Island of Riems, Germany
| | - Inês Vitoriano
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Public Health England, Porton Down, Salisbury SP4 0JG, UK
| | - Christopher H Logue
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Public Health England, Porton Down, Salisbury SP4 0JG, UK
| | - Jan Peter Boettcher
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Robert Koch Institute, 13353 Berlin, Germany
| | - Elisa Pallasch
- Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, 20359 Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, and Marburg, Germany
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
| | - Andreas Sachse
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Robert Koch Institute, 13353 Berlin, Germany
| | - Amadou Bah
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Swiss Tropical and Public Health Institute, 4051 Basel, Switzerland
| | - Katja Nitzsche
- German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, and Marburg, Germany
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
| | - Eeva Kuisma
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Public Health England, Porton Down, Salisbury SP4 0JG, UK
| | - Janine Michel
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Robert Koch Institute, 13353 Berlin, Germany
| | - Tobias Holm
- Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, 20359 Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, and Marburg, Germany
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
| | - Elsa-Gayle Zekeng
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
| | - Isabel García-Dorival
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Institute of Infection and Global Health, University of Liverpool, Liverpool L69 7BE, UK
| | - Roman Wölfel
- German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, and Marburg, Germany
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Bundeswehr Institute of Microbiology, 80937 Munich, Germany
| | - Kilian Stoecker
- German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, and Marburg, Germany
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Bundeswehr Institute of Microbiology, 80937 Munich, Germany
| | - Erna Fleischmann
- German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, and Marburg, Germany
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Bundeswehr Institute of Microbiology, 80937 Munich, Germany
| | - Thomas Strecker
- German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, and Marburg, Germany
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Institute of Virology, Philipps University, 35043 Marburg, Germany
| | - Antonino Di Caro
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- National Institute for Infectious Diseases 'Lazzaro Spallanzani', 00149 Rome, Italy
| | - Tatjana Avšič-Županc
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Andreas Kurth
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Robert Koch Institute, 13353 Berlin, Germany
| | - Silvia Meschi
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- National Institute for Infectious Diseases 'Lazzaro Spallanzani', 00149 Rome, Italy
| | - Stephane Mély
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Laboratoire P4-Jean Mérieux, US003 INSERM, 69365 Lyon, France
| | - Edmund Newman
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Public Health England, Porton Down, Salisbury SP4 0JG, UK
| | - Anne Bocquin
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Laboratoire P4-Jean Mérieux, US003 INSERM, 69365 Lyon, France
| | - Zoltan Kis
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- National Center for Epidemiology, Hungarian National Biosafety Laboratory, H1097 Budapest, Hungary
- European Centre for Disease Prevention and Control, 171 65 Solna, Sweden
| | - Anne Kelterbaum
- German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, and Marburg, Germany
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Institute of Virology, Philipps University, 35043 Marburg, Germany
| | - Peter Molkenthin
- German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, and Marburg, Germany
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Bundeswehr Institute of Microbiology, 80937 Munich, Germany
| | - Fabrizio Carletti
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- National Institute for Infectious Diseases 'Lazzaro Spallanzani', 00149 Rome, Italy
| | - Jasmine Portmann
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Federal Office for Civil Protection, CH-3700 Spiez, Switzerland
| | - Svenja Wolff
- German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, and Marburg, Germany
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Institute of Virology, Philipps University, 35043 Marburg, Germany
| | - Concetta Castilletti
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- National Institute for Infectious Diseases 'Lazzaro Spallanzani', 00149 Rome, Italy
| | - Gordian Schudt
- German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, and Marburg, Germany
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Institute of Virology, Philipps University, 35043 Marburg, Germany
| | - Alexandra Fizet
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, 69365 Lyon, France
| | - Lisa J Ottowell
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Public Health England, Porton Down, Salisbury SP4 0JG, UK
| | - Eva Herker
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, 20251 Hamburg, Germany
| | - Thomas Jacobs
- Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, 20359 Hamburg, Germany
| | - Birte Kretschmer
- Eurice, European Research and Project Office, 10115 Berlin, Germany
| | - Ettore Severi
- European Centre for Disease Prevention and Control, 171 65 Solna, Sweden
| | | | - Mar Lago
- Infectious Diseases Unit, Internal Medicine Service, Hospital La Paz, 28046 Madrid, Spain
| | - Anabel Negredo
- National Center of Microbiology, Institute of Health 'Carlos III', 28220 Madrid, Spain
| | - Leticia Franco
- National Center of Microbiology, Institute of Health 'Carlos III', 28220 Madrid, Spain
| | - Pedro Anda
- National Center of Microbiology, Institute of Health 'Carlos III', 28220 Madrid, Spain
| | - Stefan Schmiedel
- University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Benno Kreuels
- Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, 20359 Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, and Marburg, Germany
- University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Dominic Wichmann
- German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, and Marburg, Germany
- University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Marylyn M Addo
- German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, and Marburg, Germany
- University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Ansgar W Lohse
- German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, and Marburg, Germany
- University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | | | | | | | | | | | - Gao Xiaojiang
- Cancer and Inflammation Program, Laboratory of Experimental Immunology, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts 02139, USA
| | - Mary Carrington
- Cancer and Inflammation Program, Laboratory of Experimental Immunology, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts 02139, USA
| | - Osvaldo Miranda
- Hospital Militar Central Dr. Carlos J. Finlay, 11400 Havana, Cuba
| | - Carlos M Castro
- Hospital Militar Central Dr. Carlos J. Finlay, 11400 Havana, Cuba
| | - Martin Gabriel
- Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, 20359 Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, and Marburg, Germany
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
| | - Patrick Drury
- World Health Organization, 1211 Geneva 27, Switzerland
| | | | | | | | - N'Faly Magassouba
- Université Gamal Abdel Nasser de Conakry, CHU Donka, 2101 Conakry, Guinea
| | - Miles W Carroll
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
- Public Health England, Porton Down, Salisbury SP4 0JG, UK
| | - Stephan Günther
- Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, 20359 Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, and Marburg, Germany
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
| | - César Muñoz-Fontela
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, 20251 Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, World Health Organization Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, 20359 Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Sites Hamburg, Munich, and Marburg, Germany
- European Mobile Laboratory Consortium, Bernhard-Nocht-Institute for Tropical Medicine, D-20359 Hamburg, Germany
| |
Collapse
|
10
|
Dhanda SK, Chaudhary K, Gupta S, Brahmachari SK, Raghava GPS. A web-based resource for designing therapeutics against Ebola Virus. Sci Rep 2016; 6:24782. [PMID: 27113850 PMCID: PMC4845023 DOI: 10.1038/srep24782] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 04/05/2016] [Indexed: 11/23/2022] Open
Abstract
In this study, we describe a web-based resource, developed for assisting the scientific community in designing an effective therapeutics against the Ebola virus. Firstly, we predicted and identified experimentally validated epitopes in each of the antigens/proteins of the five known ebolaviruses. Secondly, we generated all the possible overlapping 9mer peptides from the proteins of ebolaviruses. Thirdly, conserved peptides across all the five ebolaviruses (four human pathogenic species) with no identical sequence in the human proteome, based on 1000 Genomes project, were identified. Finally, we identified peptide or epitope-based vaccine candidates that could activate both the B- and T-cell arms of the immune system. In addition, we also identified efficacious siRNAs against the mRNA transcriptome (absent in human transcriptome) of all the five ebolaviruses. It was observed that three species can potentially be targeted by a single siRNA (19mer) and 75 siRNAs can potentially target at least two species. A web server, EbolaVCR, has been developed that incorporates all the above information and useful computational tools (http://crdd.osdd.net/oscadd/ebola/).
Collapse
Affiliation(s)
- Sandeep Kumar Dhanda
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh, India
| | - Kumardeep Chaudhary
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh, India
| | - Sudheer Gupta
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh, India
| | | | - Gajendra P S Raghava
- Bioinformatics Centre, CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh, India
| |
Collapse
|
11
|
Dikhit MR, Kumar S, Vijaymahantesh, Sahoo BR, Mansuri R, Amit A, Yousuf Ansari M, Sahoo GC, Bimal S, Das P. Computational elucidation of potential antigenic CTL epitopes in Ebola virus. INFECTION GENETICS AND EVOLUTION 2015; 36:369-375. [PMID: 26462623 DOI: 10.1016/j.meegid.2015.10.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 09/02/2015] [Accepted: 10/09/2015] [Indexed: 11/28/2022]
Abstract
Cell-mediated immunity is important for the control of Ebola virus infection. We hypothesized that those HLA A0201 and HLA B40 restricted epitopes derived from Ebola virus proteins, would mount a good antigenic response. Here we employed an immunoinformatics approach to identify specific 9mer amino acid which may be capable of inducing a robust cell-mediated immune response in humans. We identified a set of 28 epitopes that had no homologs in humans. Specifically, the epitopes derived from NP, RdRp, GP and VP40 share population coverage of 93.40%, 84.15%, 74.94% and 77.12%, respectively. Based on the other HLA binding specificity and population coverage, seven novel promiscuous epitopes were identified. These 7 promiscuous epitopes from NP, RdRp and GP were found to have world-wide population coverage of more than 95% indicating their potential significance as useful candidates for vaccine design. Epitope conservancy analysis also suggested that most of the peptides are highly conserved (100%) in other virulent Ebola strain (Mayinga-76, Kikwit-95 and Makona-G3816- 2014) and can therefore be further investigated for their immunological relevance and usefulness as vaccine candidates.
Collapse
Affiliation(s)
- Manas R Dikhit
- Department of Bioinformatics, Rajendra Memorial Research Institute of Medical Sciences, Patna 800007, India
| | - Santosh Kumar
- Department of Biotechnology and Department of Pharmacoinformatics, National Institutes of Pharmaceutical Education and Research, Hajipur 844102, India
| | - Vijaymahantesh
- Department of Biotechnology and Department of Pharmacoinformatics, National Institutes of Pharmaceutical Education and Research, Hajipur 844102, India; Division of Immunology, Rajendra Memorial Research Institute of Medical Sciences, Patna 800007, India
| | - Bikash R Sahoo
- Institute for Protein Research, Osaka University, Suita 5650871, Japan
| | - Rani Mansuri
- Department of Bioinformatics, Rajendra Memorial Research Institute of Medical Sciences, Patna 800007, India; Department of Biotechnology and Department of Pharmacoinformatics, National Institutes of Pharmaceutical Education and Research, Hajipur 844102, India
| | - Ajay Amit
- Division of Immunology, Rajendra Memorial Research Institute of Medical Sciences, Patna 800007, India
| | - Md Yousuf Ansari
- Department of Bioinformatics, Rajendra Memorial Research Institute of Medical Sciences, Patna 800007, India; Department of Biotechnology and Department of Pharmacoinformatics, National Institutes of Pharmaceutical Education and Research, Hajipur 844102, India
| | - Ganesh C Sahoo
- Department of Bioinformatics, Rajendra Memorial Research Institute of Medical Sciences, Patna 800007, India
| | - Sanjiva Bimal
- Division of Immunology, Rajendra Memorial Research Institute of Medical Sciences, Patna 800007, India
| | - Pradeep Das
- Dept. of Molecular Parasitology, Rajendra Memorial Research Institute of Medical Sciences, Patna 800007, India.
| |
Collapse
|
12
|
Bradfute SB, Warfield KL, Bray M. Mouse models for filovirus infections. Viruses 2012; 4:1477-508. [PMID: 23170168 PMCID: PMC3499815 DOI: 10.3390/v4091477] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 08/27/2012] [Accepted: 08/27/2012] [Indexed: 12/21/2022] Open
Abstract
The filoviruses marburg- and ebolaviruses can cause severe hemorrhagic fever (HF) in humans and nonhuman primates. Because many cases have occurred in geographical areas lacking a medical research infrastructure, most studies of the pathogenesis of filoviral HF, and all efforts to develop drugs and vaccines, have been carried out in biocontainment laboratories in non-endemic countries, using nonhuman primates (NHPs), guinea pigs and mice as animal models. NHPs appear to closely mirror filoviral HF in humans (based on limited clinical data), but only small numbers may be used in carefully regulated experiments; much research is therefore done in rodents. Because of their availability in large numbers and the existence of a wealth of reagents for biochemical and immunological testing, mice have become the preferred small animal model for filovirus research. Since the first experiments following the initial 1967 marburgvirus outbreak, wild-type or mouse-adapted viruses have been tested in immunocompetent or immunodeficient mice. In this paper, we review how these types of studies have been used to investigate the pathogenesis of filoviral disease, identify immune responses to infection and evaluate antiviral drugs and vaccines. We also discuss the strengths and weaknesses of murine models for filovirus research, and identify important questions for further study.
Collapse
Affiliation(s)
- Steven B. Bradfute
- Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, NM 87131, USA
- Author to whom correspondence should be addressed; ; Tel.: +1-505-272-1433; Fax: +1-505-272-6995
| | - Kelly L. Warfield
- Vaccine Development, Integrated Biotherapeutics, Inc., Gaithersburg, MD 20878, USA;
| | - Mike Bray
- Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA;
| |
Collapse
|
13
|
Wu S, Yu T, Song X, Yi S, Hou L, Chen W. Prediction and identification of mouse cytotoxic T lymphocyte epitopes in Ebola virus glycoproteins. Virol J 2012; 9:111. [PMID: 22695180 PMCID: PMC3411508 DOI: 10.1186/1743-422x-9-111] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 06/13/2012] [Indexed: 11/10/2022] Open
Abstract
Background Ebola viruses (EBOVs) cause severe hemorrhagic fever with a high mortality rate. At present, there are no licensed vaccines or efficient therapies to combat EBOV infection. Previous studies have shown that both humoral and cellular immune responses are crucial for controlling Ebola infection. CD8+ T cells play an important role in mediating vaccine-induced protective immunity. The objective of this study was to identify H-2d-specific T cell epitopes in EBOV glycoproteins (GPs). Results Computer-assisted algorithms were used to predict H-2d-specific T cell epitopes in two species of EBOV (Sudan and Zaire) GP. The predicted peptides were synthesized and identified in BALB/c mice immunized with replication-deficient adenovirus vectors expressing the EBOV GP. Enzyme-linked immunospot assays and intracellular cytokine staining showed that the peptides RPHTPQFLF (Sudan EBOV), GPCAGDFAF and LYDRLASTV (Zaire EBOV) could stimulate splenoctyes in immunized mice to produce large amounts of interferon-gamma. Conclusion Three peptides within the GPs of two EBOV strains were identified as T cell epitopes. The identification of these epitopes should facilitate the evaluation of vaccines based on the Ebola virus glycoprotein in a BALB/c mouse model.
Collapse
Affiliation(s)
- Shipo Wu
- State Key Laboratory of Pathogens and Biosecurity, Beijing Institute of Microbiology and Epidemiology, No.20 Dongdajie Street, Fengtai district, Beijing 100071, People's Republic of China
| | | | | | | | | | | |
Collapse
|
14
|
Protective role of cytotoxic T lymphocytes in filovirus hemorrhagic fever. J Biomed Biotechnol 2011; 2011:984241. [PMID: 22253531 PMCID: PMC3255346 DOI: 10.1155/2011/984241] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Accepted: 10/21/2011] [Indexed: 11/18/2022] Open
Abstract
Infection with many emerging viruses, such as the hemorrhagic fever disease caused by the filoviruses, Marburg (MARV), and Ebola virus (EBOV), leaves the host with a short timeframe in which to mouse a protective immune response. In lethal cases, uncontrolled viral replication and virus-induced immune dysregulation are too severe to overcome, and mortality is generally associated with a lack of notable immune responses. Vaccination studies in animals have demonstrated an association of IgG and neutralizing antibody responses against the protective glycoprotein antigen with survival from lethal challenge. More recently, studies in animal models of filovirus hemorrhagic fever have established that induction of a strong filovirus-specific cytotoxic T lymphocyte (CTL) response can facilitate complete viral clearance. In this review, we describe assays used to discover CTL responses after vaccination or live filovirus infection in both animal models and human clinical trials. Unfortunately, little data regarding CTL responses have been collected from infected human survivors, primarily due to the low frequency of disease and the inability to perform these studies in the field. Advancements in assays and technologies may allow these studies to occur during future outbreaks.
Collapse
|
15
|
Tsuda Y, Caposio P, Parkins CJ, Botto S, Messaoudi I, Cicin-Sain L, Feldmann H, Jarvis MA. A replicating cytomegalovirus-based vaccine encoding a single Ebola virus nucleoprotein CTL epitope confers protection against Ebola virus. PLoS Negl Trop Dis 2011; 5:e1275. [PMID: 21858240 PMCID: PMC3153429 DOI: 10.1371/journal.pntd.0001275] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Accepted: 06/29/2011] [Indexed: 12/22/2022] Open
Abstract
Background Human outbreaks of Ebola virus (EBOV) are a serious human health concern in Central Africa. Great apes (gorillas/chimpanzees) are an important source of EBOV transmission to humans due to increased hunting of wildlife including the ‘bush-meat’ trade. Cytomegalovirus (CMV) is an highly immunogenic virus that has shown recent utility as a vaccine platform. CMV-based vaccines also have the unique potential to re-infect and disseminate through target populations regardless of prior CMV immunity, which may be ideal for achieving high vaccine coverage in inaccessible populations such as great apes. Methodology/Principal Findings We hypothesize that a vaccine strategy using CMV-based vectors expressing EBOV antigens may be ideally suited for use in inaccessible wildlife populations. To establish a ‘proof-of-concept’ for CMV-based vaccines against EBOV, we constructed a mouse CMV (MCMV) vector expressing a CD8+ T cell epitope from the nucleoprotein (NP) of Zaire ebolavirus (ZEBOV) (MCMV/ZEBOV-NPCTL). MCMV/ZEBOV-NPCTL induced high levels of long-lasting (>8 months) CD8+ T cells against ZEBOV NP in mice. Importantly, all vaccinated animals were protected against lethal ZEBOV challenge. Low levels of anti-ZEBOV antibodies were only sporadically detected in vaccinated animals prior to ZEBOV challenge suggesting a role, at least in part, for T cells in protection. Conclusions/Significance This study demonstrates the ability of a CMV-based vaccine approach to protect against an highly virulent human pathogen, and supports the potential for ‘disseminating’ CMV-based EBOV vaccines to prevent EBOV transmission in wildlife populations. Human outbreaks of hemorrhagic disease caused by Ebola virus (EBOV) are a serious health concern in Central Africa. Great apes (gorillas/chimpanzees) are an important source of EBOV transmission to humans. Candidate EBOV vaccines do not spread from the initial vaccinee. In addition to being highly immunogenic, vaccines based on the cytomegalovirus (CMV) platform have the unique potential to re-infect and disseminate through target populations. To explore the utility of CMV-based vaccines against EBOV, we constructed a mouse CMV (MCMV) vector expressing a region of nucleoprotein (NP) of Zaire ebolavirus (ZEBOV) (MCMV/ZEBOV-NPCTL). MCMV/ZEBOV-NPCTL induced high levels of long-lasting CD8+ T cells against ZEBOV NP in mice. Importantly, all vaccinated animals were protected against lethal ZEBOV challenge. The absence of ZEBOV neutralizing and only low, sporadic levels of total anti-ZEBOV IgG antibodies in protected animals prior to ZEBOV challenge indicate a role, albeit perhaps not exclusive, for CD8+ T cells in mediating protection. This study demonstrates the ability of a CMV-based vaccine approach to protect against ZEBOV, and provides a ‘proof-of-concept’ for the potential for a ‘disseminating’ CMV-based EBOV vaccine to prevent EBOV transmission in wild animal populations.
Collapse
Affiliation(s)
- Yoshimi Tsuda
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Patrizia Caposio
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Christopher J. Parkins
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Sara Botto
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Ilhem Messaoudi
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Luka Cicin-Sain
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Michael A. Jarvis
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, United States of America
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
- * E-mail:
| |
Collapse
|
16
|
Abstract
Ebolavirus causes severe hemorrhagic fever, with case fatality rates as high as 90%. Currently, no licensed vaccine is available against Ebolavirus. We previously generated a replication-deficient, biologically contained Ebolavirus, EbolaDeltaVP30, which lacks the essential VP30 gene, grows only in cells stably expressing this gene product, and is genetically stable. Here, we evaluated the vaccine potential of EbolaDeltaVP30. First, we demonstrated its safety in STAT-1-knockout mice, a susceptible animal model for Ebolavirus infection. We then tested its protective efficacy in two animal models, mice and guinea pigs. Mice immunized twice with EbolaDeltaVP30 were protected from a lethal infection of mouse-adapted Ebolavirus. Virus titers in the serum of vaccinated mice were significantly lower than those in nonvaccinated mice. Protection of mice immunized with EbolaDeltaVP30 was associated with a high antibody response to the Ebolavirus glycoprotein and the generation of an Ebolavirus NP-specific CD8(+) T-cell response. Guinea pigs immunized twice with EbolaDeltaVP30 were also protected from a lethal infection of guinea pig-adapted Ebolavirus. Our study demonstrates the potential of the EbolaDeltaVP30 virus as a new vaccine platform.
Collapse
|
17
|
Warfield K, Posten N, Swenson D, Olinger G, Esposito D, Gillette W, Hopkins R, Costantino J, Panchal R, Hartley J, Aman M, Bavari S. Filovirus‐Like Particles Produced in Insect Cells: Immunogenicity and Protection in Rodents. J Infect Dis 2007; 196 Suppl 2:S421-9. [DOI: 10.1086/520612] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
18
|
Wang L, Figueredo J, Calcedo R, Lin J, Wilson JM. Cross-presentation of adeno-associated virus serotype 2 capsids activates cytotoxic T cells but does not render hepatocytes effective cytolytic targets. Hum Gene Ther 2007; 18:185-94. [PMID: 17324107 DOI: 10.1089/hum.2007.001] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Liver toxicity observed in a clinical trial of adeno-associated virus serotype 2 (AAV2) delivered systemically to patients with hemophilia was ascribed to killing of vector-transduced hepatocytes by capsid-specific T cells. This study evaluated the biology of T cell activation in response to AAV capsids in murine models. CD8(+) T cell epitopes were mapped to capsids from AAV2, AAV7, and AAV8. A tetramer generated in response to a dominant capsid epitope in BALB/c mice was shared between these AAV serotypes. Administration of AAV2 vector resulted in the activation of capsid-specific CD8(+) T cells as evidenced by binding to tetramer and production of capsid-induced interferon-gamma expression this was not observed with the AAV7 and AAV8 vectors. CD8(+) T cells specific to AAV2 capsids demonstrate functional cytolytic activity in vivo to peptide-loaded target cells. The frequency of capsid-specific T cells was much higher in liver than in blood or spleen. The performance of liver-directed AAV-mediated gene transfer was not diminished in animals with high levels of pre-existing capsid-specific T cells. We conclude that cross-presentation of AAV capsids does result in activation of cytotoxic T lymphocytes (CTLs) in a serotype-specific manner; however, there is no evidence that vector-transduced hepatocytes are targets for CTL effector activity.
Collapse
Affiliation(s)
- Lili Wang
- Gene Therapy Program, Department of Pathology and Laboratory Medicine, Division of Transfusion Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
19
|
Sundar K, Boesen A, Coico R. Computational prediction and identification of HLA-A2.1-specific Ebola virus CTL epitopes. Virology 2006; 360:257-63. [PMID: 17123567 DOI: 10.1016/j.virol.2006.09.042] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2006] [Revised: 07/05/2006] [Accepted: 09/27/2006] [Indexed: 11/24/2022]
Abstract
Ebola virus (EBOV) is known to cause a severe hemorrhagic fever resulting in high mortality. Although the precise host defense mechanism(s) that afford protection against EBOV is not completely understood, T cell-mediated immune responses is believed to play a pivotal role in controlling virus replication and EBOV infection. There have been no reports on mapping of MHC Class I-binding CTL epitopes for EBOV till to date. In this study, we identified five HLA-A2-binding 9-mer peptides of EBOV nucleoprotein (NP) using computer-assisted algorithm. The peptides were synthesized and examined for their ability to bind to MHC class I molecules using a flow cytometry based MHC stabilization assay. Three of the EBOV-NP peptides tested (FLSFASLFL, RLMRTNFLI and KLTEAITAA) stabilized HLA-A2. The ability of the HLA-A2-binding EBOV-NP peptides to generate peptide-specific CTLs was evaluated in HLA-A2.1 transgenic mice. Epitope-specific CTL responses were confirmed by cytotoxic assays against peptide-pulsed target cells and interferon-gamma ELISPOT assay. Each of the EBOV-NP peptides induced CTL responses in HLA-A2-transgenic mice. Interestingly, all the three peptides were conserved in three different strains of Ebola (Zaire and Reston and Sudan). Taken together, these findings provide direct evidence for the existence of EBOV-derived NP epitopes that may be useful in the development of protective immunogens for this hemorrhagic virus.
Collapse
Affiliation(s)
- Krishnan Sundar
- Department of Microbiology and Immunology, City University of New York Medical School, New York, NY 10031, USA
| | | | | |
Collapse
|
20
|
Vandenberghe LH, Wang L, Somanathan S, Zhi Y, Figueredo J, Calcedo R, Sanmiguel J, Desai RA, Chen CS, Johnston J, Grant RL, Gao G, Wilson JM. Heparin binding directs activation of T cells against adeno-associated virus serotype 2 capsid. Nat Med 2006; 12:967-71. [PMID: 16845388 DOI: 10.1038/nm1445] [Citation(s) in RCA: 166] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2006] [Accepted: 06/12/2006] [Indexed: 01/02/2023]
Abstract
Activation of T cells to the capsid of adeno-associated virus (AAV) serotype 2 vectors has been implicated in liver toxicity in a recent human gene therapy trial of hemophilia B. To further investigate this kind of toxicity, we evaluated T-cell responses to AAV capsids after intramuscular injection of vectors into mice and nonhuman primates. High levels of T cells specific to capsids of vectors based on AAV2 and a phylogenetically related AAV variant were detected. Vectors from other AAV clades such as AAV8 (ref. 3), however, did not lead to activation of capsid-specific T cells. Through the generation of AAV2-AAV8 hybrids and the creation of site-directed mutations, we mapped the domain that directs the activation of T cells to the RXXR motif on VP3, which was previously shown to confer binding of the virion to heparan sulfate proteoglycan (HSPG). Evaluation of natural and engineered AAV variants showed direct correlations between heparin binding, uptake into human dendritic cells (DCs) and activation of capsid-specific T cells. The role of heparin binding in the activation of CD8(+) T cells may be useful in modulating the immunogenicity of antigens and improving the safety profile of existing AAV vectors for gene therapy.
Collapse
Affiliation(s)
- Luk H Vandenberghe
- Gene Therapy Program, Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, 125 S. 31st Street, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Olinger GG, Bailey MA, Dye JM, Bakken R, Kuehne A, Kondig J, Wilson J, Hogan RJ, Hart MK. Protective cytotoxic T-cell responses induced by venezuelan equine encephalitis virus replicons expressing Ebola virus proteins. J Virol 2006; 79:14189-96. [PMID: 16254354 PMCID: PMC1280180 DOI: 10.1128/jvi.79.22.14189-14196.2005] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Infection with Ebola virus causes a severe disease accompanied by high mortality rates, and there are no licensed vaccines or therapies available for human use. Filovirus vaccine research efforts still need to determine the roles of humoral and cell-mediated immune responses in protection from Ebola virus infection. Previous studies indicated that exposure to Ebola virus proteins expressed from packaged Venezuelan equine encephalitis virus replicons elicited protective immunity in mice and that antibody-mediated protection could only be demonstrated after vaccination against the glycoprotein. In this study, the murine CD8(+) T-cell responses to six Ebola virus proteins were examined. CD8(+) T cells specific for Ebola virus glycoprotein, nucleoprotein, and viral proteins (VP24, VP30, VP35, and VP40) were identified by intracellular cytokine assays using splenocytes from vaccinated mice. The cells were expanded by restimulation with peptides and demonstrated cytolytic activity. Adoptive transfer of the CD8(+) cytotoxic T cells protected filovirus naïve mice from challenge with Ebola virus. These data support a role for CD8(+) cytotoxic T cells as part of a protective mechanism induced by vaccination against six Ebola virus proteins and provide additional evidence that cytotoxic T-cell responses can contribute to protection from filovirus infections.
Collapse
Affiliation(s)
- Gene G Olinger
- United States Army Medical Research Institute of Infectious Diseases, Division of Virology, 1425 Porter Street, Frederick, MD 21702-5011, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Boesen A, Sundar K, Coico R. Lassa fever virus peptides predicted by computational analysis induce epitope-specific cytotoxic-T-lymphocyte responses in HLA-A2.1 transgenic mice. CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY 2005; 12:1223-30. [PMID: 16210487 PMCID: PMC1247823 DOI: 10.1128/cdli.12.10.1223-1230.2005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2005] [Revised: 05/17/2005] [Accepted: 06/16/2005] [Indexed: 11/20/2022]
Abstract
Lassa fever is a hemorrhagic disease caused by Lassa fever virus (LV). Although the precise host defense mechanism(s) that affords protection against LV is not completely understood, cellular immunity mediated by cytotoxic T lymphocytes (CTLs) plays a pivotal role in controlling viral replication and LV infection. To date, there have been no reports mapping major histocompatibility complex (MHC) class I-binding CTL epitopes for LV. Using computer-assisted algorithms, we identified five HLA-A2.1-binding peptides of LV glycoprotein (GP) and two peptides from LV nucleoprotein (NP). Synthesized peptides were examined for their ability to bind to MHC class I molecules using a flow cytometric assay that measures peptide stabilization of class I. Three of the LV-GP peptides tested (LLGTFTWTL, SLYKGVYEL, and YLISIFLHL) stabilized HLA-A2. The LV-NP peptides tested failed to stabilize this HLA-A2. We then investigated the ability of the HLA-A2-binding LV-GP peptides to generate peptide-specific CTLs in HLA-A2.1 transgenic mice. Functional assays used to confirm CTL activation included gamma interferon enzyme-linked immunospot (ELISPOT) assays and intracellular cytokine staining of CD8+ T cells from peptide-primed mice. CTL assays were also performed to verify the cytolytic activity of peptide-pulsed target cells. Each of the LV-GP peptides induced CTL responses in HLA-A2-transgenic mice. MHC class I tetramers prepared using one LV-GP peptide that showed the highest cytolytic index (LLGTFTWTL) confirmed that peptide-binding CD8+ T cells were present in pooled lymphocytes harvested from peptide-primed mice. These findings provide direct evidence for the existence of LV-derived GP epitopes that may be useful in the development of protective immunogens for this hemorrhagic virus.
Collapse
Affiliation(s)
- Agnieszka Boesen
- Department of Microbiology and Immunology, City University of New York Medical School, New York, New York 10031, USA
| | | | | |
Collapse
|
23
|
Warfield KL, Olinger G, Deal EM, Swenson DL, Bailey M, Negley DL, Hart MK, Bavari S. Induction of Humoral and CD8+ T Cell Responses Are Required for Protection against Lethal Ebola Virus Infection. THE JOURNAL OF IMMUNOLOGY 2005; 175:1184-91. [PMID: 16002721 DOI: 10.4049/jimmunol.175.2.1184] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Ebola virus (EBOV)-like particles (eVLP), composed of the EBOV glycoprotein and matrix viral protein (VP)40 with a lipid membrane, are a highly efficacious method of immunization against EBOV infection. The exact requirements for immunity against EBOV infection are poorly defined at this time. The goal of this work was to determine the requirements for EBOV immunity following eVLP vaccination. Vaccination of BALB/c or C57BL/6 mice with eVLPs in conjunction with QS-21 adjuvant resulted in mixed IgG subclass responses, a Th1-like memory cytokine response, and protection from lethal EBOV challenge. Further, this vaccination schedule led to the generation of both CD4(+) and CD8(+) IFN-gamma(+) T cells recognizing specific peptides within glycoprotein and VP40. The transfer of both serum and splenocytes, but not serum or splenocytes alone, from eVLP-vaccinated mice conferred protection against lethal EBOV infection in these studies. B cells were required for eVLP-mediated immunity to EBOV because B cell-deficient mice vaccinated with eVLPs were not protected from lethal EBOV challenge. We also found that CD8(+), but not CD4(+), T cells are absolutely required for eVLP-mediated protection against EBOV infection. Further, eVLP-induced protective mechanisms were perforin-independent, but IFN-gamma-dependent. Taken together, both EBOV-specific humoral and cytotoxic CD8(+) T cell responses are critical to mediate protection against filoviruses following eVLP vaccination.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Amino Acid Sequence
- Animals
- Antibodies, Viral/biosynthesis
- Antibodies, Viral/physiology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/virology
- Cells, Cultured
- Ebolavirus/immunology
- Epitopes, T-Lymphocyte/immunology
- Female
- Hemorrhagic Fever, Ebola/immunology
- Hemorrhagic Fever, Ebola/mortality
- Hemorrhagic Fever, Ebola/prevention & control
- Interferon-gamma/biosynthesis
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Molecular Sequence Data
- Saponins/administration & dosage
- Saponins/immunology
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/immunology
- Viral Envelope Proteins/immunology
- Viral Matrix Proteins/immunology
- Viral Vaccines/administration & dosage
- Viral Vaccines/immunology
- Virion/immunology
Collapse
Affiliation(s)
- Kelly L Warfield
- U.S. Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Frederick, MD 21702, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Gupta M, Greer P, Mahanty S, Shieh WJ, Zaki SR, Ahmed R, Rollin PE. CD8-Mediated Protection against Ebola Virus Infection Is Perforin Dependent. THE JOURNAL OF IMMUNOLOGY 2005; 174:4198-202. [DOI: 10.4049/jimmunol.174.7.4198] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|