1
|
Mihaylova NM, Manoylov IK, Nikolova MH, Prechl J, Tchorbanov AI. DNA and protein-generated chimeric molecules for delivery of influenza viral epitopes in mouse and humanized NSG transfer models. Hum Vaccin Immunother 2024; 20:2292381. [PMID: 38193304 PMCID: PMC10793685 DOI: 10.1080/21645515.2023.2292381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 12/05/2023] [Indexed: 01/10/2024] Open
Abstract
Purified subunit viral antigens are weakly immunogenic and stimulate only the antibody but not the T cell-mediated immune response. An alternative approach to inducing protective immunity with small viral peptides may be the targeting of viral epitopes to immunocompetent cells by DNA and protein-engineered vaccines. This review will focus on DNA and protein-generated chimeric molecules carrying engineered fragments specific for activating cell surface co-receptors for inducing protective antiviral immunity. Adjuvanted protein-based vaccine or DNA constructs encoding simultaneously T- and B-cell peptide epitopes from influenza viral hemagglutinin, and scFvs specific for costimulatory immune cell receptors may induce a significant increase of anti-influenza antibody levels and strong CTL activity against virus-infected cells in a manner that mimics the natural infection. Here we summarize the development of several DNA and protein chimeric constructs carrying influenza virus HA317-41 fragment. The generated engineered molecules were used for immunization in intact murine and experimentally humanized NSG mouse models.
Collapse
Affiliation(s)
- Nikolina M. Mihaylova
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Iliyan K. Manoylov
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Maria H. Nikolova
- National Reference Laboratory of Immunology, National Center of Infectious and Parasitic Diseases, Sofia, Bulgaria
| | | | - Andrey I. Tchorbanov
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
- National Institute of Immunology, Sofia, Bulgaria
| |
Collapse
|
2
|
Ding X, Zhao F, Liu Z, Yao J, Yu H, Zhang X. Original antigenic sin: A potential double-edged effect for vaccine improvement. Biomed Pharmacother 2024; 178:117187. [PMID: 39084082 DOI: 10.1016/j.biopha.2024.117187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024] Open
Abstract
Original antigenic sin (OAS) influences the immune response to subsequent infections with related variants following initial pathogen exposure. This phenomenon is characterized by cross-reactivity, which, although it may worsen infections, also provides a degree of protection against immune evasion caused by variations. This paradox complicates the development of creating universal vaccinations, as they frequently show diminished effectiveness against these emerging variants. This review aims to elucidate the diverse impacts of OAS on the immune response to various infections, emphasizing the complicated balance between beneficial and harmful outcomes. Moreover, we evaluate the influence of adjuvants and other variables on the extent of OAS, hence affecting the effectiveness of vaccines. Understanding the mechanisms of OAS that cause persistent infections and evasion of the immune system is crucial for the developing innovative vaccines. And it has significant potential for clinical applications.
Collapse
Affiliation(s)
- Xuan Ding
- MOE Key Lab of Rare Pediatric Diseases &Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang 421001, PR China
| | - Feijun Zhao
- MOE Key Lab of Rare Pediatric Diseases &Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang 421001, PR China; Laboratory Medicine Center, the First Affiliated Hospital of University of South ChinaHengyang 421001, PR China
| | - Zhaoping Liu
- MOE Key Lab of Rare Pediatric Diseases &Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang 421001, PR China
| | - Jiangchen Yao
- MOE Key Lab of Rare Pediatric Diseases &Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang 421001, PR China
| | - Han Yu
- MOE Key Lab of Rare Pediatric Diseases &Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang 421001, PR China
| | - Xiaohong Zhang
- MOE Key Lab of Rare Pediatric Diseases &Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang 421001, PR China.
| |
Collapse
|
3
|
Amoah S, Cao W, Sayedahmed EE, Wang Y, Kumar A, Mishina M, Eddins DJ, Wang WC, Burroughs M, Sheth M, Lee J, Shieh WJ, Ray SD, Bohannon CD, Ranjan P, Sharma SD, Hoehner J, Arthur RA, Gangappa S, Wakamatsu N, Johnston HR, Pohl J, Mittal SK, Sambhara S. The frequency and function of nucleoprotein-specific CD8 + T cells are critical for heterosubtypic immunity against influenza virus infection. J Virol 2024; 98:e0071124. [PMID: 39082839 PMCID: PMC11334528 DOI: 10.1128/jvi.00711-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 06/27/2024] [Indexed: 08/21/2024] Open
Abstract
Cytotoxic T lymphocytes (CTLs) mediate host defense against viral and intracellular bacterial infections and tumors. However, the magnitude of CTL response and their function needed to confer heterosubtypic immunity against influenza virus infection are unknown. We addressed the role of CD8+ T cells in the absence of any cross-reactive antibody responses to influenza viral proteins using an adenoviral vector expressing a 9mer amino acid sequence recognized by CD8+ T cells. Our results indicate that both CD8+ T cell frequency and function are crucial for heterosubtypic immunity. Low morbidity, lower viral lung titers, low to minimal lung pathology, and better survival upon heterosubtypic virus challenge correlated with the increased frequency of NP-specific CTLs. NP-CD8+ T cells induced by differential infection doses displayed distinct RNA transcriptome profiles and functional properties. CD8+ T cells induced by a high dose of influenza virus secreted significantly higher levels of IFN-γ and exhibited higher levels of cytotoxic function. The mice that received NP-CD8+ T cells from the high-dose virus recipients through adoptive transfer had lower viral titers following viral challenge than those induced by the low dose of virus, suggesting differential cellular programming by antigen dose. Enhanced NP-CD8+ T-cell functions induced by a higher dose of influenza virus strongly correlated with the increased expression of cellular and metabolic genes, indicating a shift to a more glycolytic metabolic phenotype. These findings have implications for developing effective T cell vaccines against infectious diseases and cancer. IMPORTANCE Cytotoxic T lymphocytes (CTLs) are an important component of the adaptive immune system that clears virus-infected cells or tumor cells. Hence, developing next-generation vaccines that induce or recall CTL responses against cancer and infectious diseases is crucial. However, it is not clear if the frequency, function, or both are essential in conferring protection, as in the case of influenza. In this study, we demonstrate that both CTL frequency and function are crucial for providing heterosubtypic immunity to influenza by utilizing an Ad-viral vector expressing a CD8 epitope only to rule out the role of antibodies, single-cell RNA-seq analysis, as well as adoptive transfer experiments. Our findings have implications for developing T cell vaccines against infectious diseases and cancer.
Collapse
Affiliation(s)
- Samuel Amoah
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Weiping Cao
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Ekramy E. Sayedahmed
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, USA
| | - Yuanyuan Wang
- Biotechnology Core Facility Branch, Division of Scientific Resources, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Amrita Kumar
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Margarita Mishina
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Devon J. Eddins
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Wen-Chien Wang
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, USA
| | - Mark Burroughs
- Biotechnology Core Facility Branch, Division of Scientific Resources, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Mili Sheth
- Biotechnology Core Facility Branch, Division of Scientific Resources, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Justin Lee
- Biotechnology Core Facility Branch, Division of Scientific Resources, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Wun-Ju Shieh
- Division of High Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Sean D. Ray
- Department of Biology, Georgia State University, Atlanta, Georgia, USA
| | - Caitlin D. Bohannon
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Priya Ranjan
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Suresh D. Sharma
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Jessica Hoehner
- Emory Integrated Computational Core, Emory Integrated Core Facilities, Emory University, Atlanta, Georgia, USA
| | - Robert A. Arthur
- Emory Integrated Computational Core, Emory Integrated Core Facilities, Emory University, Atlanta, Georgia, USA
| | - Shivaprakash Gangappa
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Nobuko Wakamatsu
- Indiana Animal Disease Diagnostic Laboratory, Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, USA
| | - H. Richard Johnston
- Department of Human Genetics, Emory University School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Jan Pohl
- Biotechnology Core Facility Branch, Division of Scientific Resources, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Suresh K. Mittal
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, USA
| | - Suryaprakash Sambhara
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
4
|
Manoutcharian K, Gevorkian G. Are we getting closer to a successful neoantigen cancer vaccine? Mol Aspects Med 2024; 96:101254. [PMID: 38354548 DOI: 10.1016/j.mam.2024.101254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/16/2024]
Abstract
Although significant advances in immunotherapy have revolutionized the treatment of many cancer types over the past decade, the field of vaccine therapy, an important component of cancer immunotherapy, despite decades-long intense efforts, is still transmitting signals of promises and awaiting strong data on efficacy to proceed with regulatory approval. The field of cancer vaccines faces standard challenges, such as tumor-induced immunosuppression, immune response in inhibitory tumor microenvironment (TME), intratumor heterogeneity (ITH), permanently evolving cancer mutational landscape leading to neoantigens, and less known obstacles: neoantigen gain/loss upon immunotherapy, the timing and speed of appearance of neoantigens and responding T cell clonotypes and possible involvement of immune interference/heterologous immunity, in the complex interplay between evolving tumor epitopes and the immune system. In this review, we discuss some key issues related to challenges hampering the development of cancer vaccines, along with the current approaches focusing on neoantigens. We summarize currently well-known ideas/rationales, thus revealing the need for alternative vaccine approaches. Such a discussion should stimulate vaccine researchers to apply out-of-box, unconventional thinking in search of new avenues to deal with critical, often yet unaddressed challenges on the road to a new generation of therapeutics and vaccines.
Collapse
Affiliation(s)
- Karen Manoutcharian
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), CDMX, Apartado Postal 70228, Cuidad Universitaria, Mexico DF, CP, 04510, Mexico.
| | - Goar Gevorkian
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), CDMX, Apartado Postal 70228, Cuidad Universitaria, Mexico DF, CP, 04510, Mexico.
| |
Collapse
|
5
|
Kim SH, Kim J, Jung S, Noh JY, Kim J, Park H, Song YG, Peck KR, Park SH, Park MS, Ko JH, Song JY, Choi JY, Jung MK, Shin EC. Omicron BA.2 breakthrough infection elicits CD8 + T cell responses recognizing the spike of later Omicron subvariants. Sci Immunol 2024; 9:eade6132. [PMID: 38241400 DOI: 10.1126/sciimmunol.ade6132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 12/14/2023] [Indexed: 01/21/2024]
Abstract
Here, we examine peripheral blood memory T cell responses against the SARS-CoV-2 BA.4/BA.5 variant spike among vaccinated individuals with or without Omicron breakthrough infections. We provide evidence supporting a lack of original antigenic sin in CD8+ T cell responses targeting the spike. We show that BNT162b2-induced memory T cells respond to the BA.4/BA.5 spike. Among individuals with BA.1/BA.2 breakthrough infections, IFN-γ-producing CD8+ T cell responses against the BA.4/BA.5 spike increased. In a subgroup with BA.2 breakthrough infections, IFN-γ-producing CD8+ T cell responses against the BA.2-mutated spike region increased and correlated directly with responses against the BA.4/BA.5 spike, indicating that BA.2 spike-specific CD8+ T cells elicited by BA.2 breakthrough infection cross-react with the BA.4/BA.5 spike. We identified CD8+ T cell epitope peptides that are present in the spike of BA.2 and BA.4/BA.5 but not the original spike. These peptides are fully conserved in the spike of now-dominant XBB lineages. Our study shows that breakthrough infection by early Omicron subvariants elicits CD8+ T cell responses that recognize epitopes within the spike of newly emerging subvariants.
Collapse
Affiliation(s)
- Sang-Hoon Kim
- Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea
| | - Jihye Kim
- Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea
| | - Sungmin Jung
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Ji Yun Noh
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul 08308, Republic of Korea
| | - Jinnam Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Heedo Park
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Young Goo Song
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Kyong Ran Peck
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Su-Hyung Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Man-Seong Park
- Department of Microbiology, Institute for Viral Diseases, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Jae-Hoon Ko
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Joon Young Song
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul 08308, Republic of Korea
| | - Jun Yong Choi
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Min Kyung Jung
- Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea
| | - Eui-Cheol Shin
- Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| |
Collapse
|
6
|
van Bergen J, Camps MG, Pardieck IN, Veerkamp D, Leung WY, Leijs AA, Myeni SK, Kikkert M, Arens R, Zondag GC, Ossendorp F. Multiantigen pan-sarbecovirus DNA vaccines generate protective T cell immune responses. JCI Insight 2023; 8:e172488. [PMID: 37707962 PMCID: PMC10721273 DOI: 10.1172/jci.insight.172488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/12/2023] [Indexed: 09/16/2023] Open
Abstract
SARS-CoV-2 is the third zoonotic coronavirus to cause a major outbreak in humans in recent years, and many more SARS-like coronaviruses with pandemic potential are circulating in several animal species. Vaccines inducing T cell immunity against broadly conserved viral antigens may protect against hospitalization and death caused by outbreaks of such viruses. We report the design and preclinical testing of 2 T cell-based pan-sarbecovirus vaccines, based on conserved regions within viral proteins of sarbecovirus isolates of human and other carrier animals, like bats and pangolins. One vaccine (CoVAX_ORF1ab) encoded antigens derived from nonstructural proteins, and the other (CoVAX_MNS) encoded antigens from structural proteins. Both multiantigen DNA vaccines contained a large set of antigens shared across sarbecoviruses and were rich in predicted and experimentally validated human T cell epitopes. In mice, the multiantigen vaccines generated both CD8+ and CD4+ T cell responses to shared epitopes. Upon encounter of full-length spike antigen, CoVAX_MNS-induced CD4+ T cells were responsible for accelerated CD8+ T cell and IgG Ab responses specific to the incoming spike, irrespective of its sarbecovirus origin. Finally, both vaccines elicited partial protection against a lethal SARS-CoV-2 challenge in human angiotensin-converting enzyme 2-transgenic mice. These results support clinical testing of these universal sarbecovirus vaccines for pandemic preparedness.
Collapse
Affiliation(s)
| | - Marcel G.M. Camps
- Department of Immunology, Leiden University Medical Centre, Leiden, Netherlands
| | - Iris N. Pardieck
- Department of Immunology, Leiden University Medical Centre, Leiden, Netherlands
| | - Dominique Veerkamp
- Department of Immunology, Leiden University Medical Centre, Leiden, Netherlands
| | - Wing Yan Leung
- Immunetune BV, Leiden, Netherlands
- Synvolux BV, Leiden, Netherlands
| | - Anouk A. Leijs
- Department of Medical Microbiology, Leiden University Medical Centre, Leiden, Netherlands
| | - Sebenzile K. Myeni
- Department of Medical Microbiology, Leiden University Medical Centre, Leiden, Netherlands
| | - Marjolein Kikkert
- Department of Medical Microbiology, Leiden University Medical Centre, Leiden, Netherlands
| | - Ramon Arens
- Department of Immunology, Leiden University Medical Centre, Leiden, Netherlands
| | - Gerben C. Zondag
- Immunetune BV, Leiden, Netherlands
- Synvolux BV, Leiden, Netherlands
| | - Ferry Ossendorp
- Department of Immunology, Leiden University Medical Centre, Leiden, Netherlands
| |
Collapse
|
7
|
Diniz MO, Maini MK, Swadling L. T cell control of SARS-CoV-2: When, which, and where? Semin Immunol 2023; 70:101828. [PMID: 37651850 DOI: 10.1016/j.smim.2023.101828] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/13/2023] [Indexed: 09/02/2023]
Abstract
Efficient immune protection against viruses such as SARS-CoV-2 requires the coordinated activity of innate immunity, B and T cells. Accumulating data point to a critical role for T cells not only in the clearance of established infection, but also for aborting viral replication independently of humoral immunity. Here we review the evidence supporting the contribution of antiviral T cells and consider which of their qualitative features favour efficient control of infection. We highlight how studies of SARS-CoV-2 and other coronaviridae in animals and humans have provided important lessons on the optimal timing (When), functionality and specificity (Which), and location (Where) of antiviral T cells. We discuss the clinical implications, particularly for the development of next-generation vaccines, and emphasise areas requiring further study.
Collapse
Affiliation(s)
- Mariana O Diniz
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London WC1E 6BT, UK
| | - Mala K Maini
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London WC1E 6BT, UK.
| | - Leo Swadling
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London WC1E 6BT, UK.
| |
Collapse
|
8
|
Ghorani E, Swanton C, Quezada SA. Cancer cell-intrinsic mechanisms driving acquired immune tolerance. Immunity 2023; 56:2270-2295. [PMID: 37820584 DOI: 10.1016/j.immuni.2023.09.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/11/2023] [Accepted: 09/11/2023] [Indexed: 10/13/2023]
Abstract
Immune evasion is a hallmark of cancer, enabling tumors to survive contact with the host immune system and evade the cycle of immune recognition and destruction. Here, we review the current understanding of the cancer cell-intrinsic factors driving immune evasion. We focus on T cells as key effectors of anti-cancer immunity and argue that cancer cells evade immune destruction by gaining control over pathways that usually serve to maintain physiological tolerance to self. Using this framework, we place recent mechanistic advances in the understanding of cancer immune evasion into broad categories of control over T cell localization, antigen recognition, and acquisition of optimal effector function. We discuss the redundancy in the pathways involved and identify knowledge gaps that must be overcome to better target immune evasion, including the need for better, routinely available tools that incorporate the growing understanding of evasion mechanisms to stratify patients for therapy and trials.
Collapse
Affiliation(s)
- Ehsan Ghorani
- Cancer Immunology and Immunotherapy Unit, Department of Surgery and Cancer, Imperial College London, London, UK; Department of Medical Oncology, Imperial College London Hospitals, London, UK.
| | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK; Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Department of Oncology, University College London Hospitals, London, UK
| | - Sergio A Quezada
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK; Cancer Immunology Unit, Research Department of Hematology, University College London Cancer Institute, London, UK.
| |
Collapse
|
9
|
Müller TR, Sekine T, Trubach D, Niessl J, Chen P, Bergman P, Blennow O, Hansson L, Mielke S, Nowak P, Vesterbacka J, Akber M, Olofsson A, Amaya Hernandez SP, Gao Y, Cai C, Söderdahl G, Smith CIE, Österborg A, Loré K, Sällberg Chen M, Ljungman P, Ljunggren HG, Karlsson AC, Saini SK, Aleman S, Buggert M. Additive effects of booster mRNA vaccination and SARS-CoV-2 Omicron infection on T cell immunity across immunocompromised states. Sci Transl Med 2023; 15:eadg9452. [PMID: 37437015 PMCID: PMC7615622 DOI: 10.1126/scitranslmed.adg9452] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 06/23/2023] [Indexed: 07/14/2023]
Abstract
Suboptimal immunity to SARS-CoV-2 mRNA vaccination has frequently been observed in individuals with various immunodeficiencies. Given the increased antibody evasion properties of emerging SARS-CoV-2 subvariants, it is necessary to assess whether other components of adaptive immunity generate resilient and protective responses against infection. We assessed T cell responses in 279 individuals, covering five different immunodeficiencies and healthy controls, before and after booster mRNA vaccination, as well as after Omicron infection in a subset of patients. We observed robust and persistent Omicron-reactive T cell responses that increased markedly upon booster vaccination and correlated directly with antibody titers across all patient groups. Poor vaccination responsiveness in immunocompromised or elderly individuals was effectively counteracted by the administration of additional vaccine doses. Functionally, Omicron-reactive T cell responses exhibited a pronounced cytotoxic profile and signs of longevity, characterized by CD45RA+ effector memory subpopulations with stem cell-like properties and increased proliferative capacity. Regardless of underlying immunodeficiency, booster-vaccinated and Omicron-infected individuals appeared protected against severe disease and exhibited enhanced and diversified T cell responses against conserved and Omicron-specific epitopes. Our findings indicate that T cells retain the ability to generate highly functional responses against newly emerging variants, even after repeated antigen exposure and a robust immunological imprint from ancestral SARS-CoV-2 mRNA vaccination.
Collapse
Affiliation(s)
- Thomas R. Müller
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Takuya Sekine
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Darya Trubach
- Department of Health Technology, Section of Experimental and Translational Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Julia Niessl
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Puran Chen
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Peter Bergman
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Laboratory Medicine, Clinical Immunology, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Ola Blennow
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Transplantation, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Huddinge, Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
| | - Lotta Hansson
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Stephan Mielke
- Department of Laboratory Medicine, Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska Comprehensive Cancer Center, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Piotr Nowak
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Huddinge, Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
- Laboratory for Molecular Infection Medicine Sweden MIMS, Umeå University, Sweden
| | - Jan Vesterbacka
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Huddinge, Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
| | - Mira Akber
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna Olofsson
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Susana Patricia Amaya Hernandez
- Department of Health Technology, Section of Experimental and Translational Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Yu Gao
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Curtis Cai
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Gunnar Söderdahl
- Department of Transplantation, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - C. I. Edvard Smith
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Laboratory Medicine, Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska Comprehensive Cancer Center, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Anders Österborg
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Karin Loré
- Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | | - Per Ljungman
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska Comprehensive Cancer Center, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Department of Medicine Huddinge, Hematology, Karolinska Institutet, Stockholm
| | - Hans-Gustaf Ljunggren
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Annika C. Karlsson
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Laboratory, Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden
| | - Sunil Kumar Saini
- Department of Health Technology, Section of Experimental and Translational Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Soo Aleman
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Huddinge, Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
| | - Marcus Buggert
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
10
|
Mosquera-Sulbaran JA, Pedreañez A, Hernandez-Fonseca JP, Hernandez-Fonseca H. Angiotensin II and dengue. Arch Virol 2023; 168:191. [PMID: 37368044 DOI: 10.1007/s00705-023-05814-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 05/09/2023] [Indexed: 06/28/2023]
Abstract
Dengue is a disease caused by a flavivirus that is transmitted principally by the bite of an Aedes aegypti mosquito and represents a major public-health problem. Many studies have been carried out to identify soluble factors that are involved in the pathogenesis of this infection. Cytokines, soluble factors, and oxidative stress have been reported to be involved in the development of severe disease. Angiotensin II (Ang II) is a hormone with the ability to induce the production of cytokines and soluble factors related to the inflammatory processes and coagulation disorders observed in dengue. However, a direct involvement of Ang II in this disease has not been demonstrated. This review primarily summarizes the pathophysiology of dengue, the role of Ang II in various diseases, and reports that are highly suggestive of the involvement of this hormone in dengue.
Collapse
Affiliation(s)
- Jesus A Mosquera-Sulbaran
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette", Facultad de Medicina, Universidad del Zulia, Maracaibo, 4001-A, Venezuela.
| | - Adriana Pedreañez
- Cátedra de Inmunología, Escuela de Bioanálisis, Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
| | - Juan Pablo Hernandez-Fonseca
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette", Facultad de Medicina, Universidad del Zulia, Maracaibo, 4001-A, Venezuela
- Servicio de Microscopia Electronica del Centro Nacional de Biotecnologia (CNB- CSIC) Madrid, Madrid, España
| | - Hugo Hernandez-Fonseca
- Department of Anatomy, Physiology and Pharmacology, School of Veterinary Medicine, Saint George's University, True Blue, West Indies, Grenada
| |
Collapse
|
11
|
Lista F, Peragallo MS, Biselli R, De Santis R, Mariotti S, Nisini R, D'Amelio R. Have Diagnostics, Therapies, and Vaccines Made the Difference in the Pandemic Evolution of COVID-19 in Comparison with "Spanish Flu"? Pathogens 2023; 12:868. [PMID: 37513715 PMCID: PMC10384375 DOI: 10.3390/pathogens12070868] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/15/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
In 1918 many countries, but not Spain, were fighting World War I. Spanish press could report about the diffusion and severity of a new infection without censorship for the first-time, so that this pandemic is commonly defined as "Spanish flu", even though Spain was not its place of origin. "Spanish flu" was one of the deadliest pandemics in history and has been frequently compared with the coronavirus disease (COVID)-19 pandemic. These pandemics share similarities, being both caused by highly variable and transmissible respiratory RNA viruses, and diversity, represented by diagnostics, therapies, and especially vaccines, which were made rapidly available for COVID-19, but not for "Spanish flu". Most comparison studies have been carried out in the first period of COVID-19, when these resources were either not yet available or their use had not long started. Conversely, we wanted to analyze the role that the advanced diagnostics, anti-viral agents, including monoclonal antibodies, and innovative COVID-19 vaccines, may have had in the pandemic containment. Early diagnosis, therapies, and anti-COVID-19 vaccines have markedly reduced the pandemic severity and mortality, thus preventing the collapse of the public health services. However, their influence on the reduction of infections and re-infections, thus on the transition from pandemic to endemic condition, appears to be of minor relevance. The high viral variability of influenza and coronavirus may probably be contained by the development of universal vaccines, which are not easy to be obtained. The only effective weapon still remains the disease prevention, to be achieved with the reduction of promiscuity between the animal reservoirs of these zoonotic diseases and humans.
Collapse
Affiliation(s)
- Florigio Lista
- Istituto di Scienze Biomediche della Difesa, Ispettorato Generale della Sanità Militare, Stato Maggiore della Difesa, 00184 Roma, Italy
| | - Mario Stefano Peragallo
- Centro Studi e Ricerche di Sanità e Veterinaria, Comando Logistico dell'Esercito, 00184 Roma, Italy
| | - Roberto Biselli
- Ispettorato Generale della Sanità Militare, Stato Maggiore della Difesa, 00184 Roma, Italy
| | - Riccardo De Santis
- Istituto di Scienze Biomediche della Difesa, Ispettorato Generale della Sanità Militare, Stato Maggiore della Difesa, 00184 Roma, Italy
- Dipartimento di Sanità Pubblica e Malattie Infettive, Sapienza, Università di Roma, 00161 Roma, Italy
| | - Sabrina Mariotti
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, 00161 Roma, Italy
| | - Roberto Nisini
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, 00161 Roma, Italy
| | - Raffaele D'Amelio
- Dipartimento di Medicina Clinica e Molecolare, Sapienza, Università di Roma, 00198 Roma, Italy
| |
Collapse
|
12
|
Aguilar-Bretones M, Fouchier RA, Koopmans MP, van Nierop GP. Impact of antigenic evolution and original antigenic sin on SARS-CoV-2 immunity. J Clin Invest 2023; 133:e162192. [PMID: 36594464 PMCID: PMC9797340 DOI: 10.1172/jci162192] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Infections with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and vaccinations targeting the spike protein (S) offer protective immunity against coronavirus disease 2019 (COVID-19). This immunity may further be shaped by cross-reactivity with common cold coronaviruses. Mutations arising in S that are associated with altered intrinsic virus properties and immune escape result in the continued circulation of SARS-CoV-2 variants. Potentially, vaccine updates will be required to protect against future variants of concern, as for influenza. To offer potent protection against future variants, these second-generation vaccines may need to redirect immunity to epitopes associated with immune escape and not merely boost immunity toward conserved domains in preimmune individuals. For influenza, efficacy of repeated vaccination is hampered by original antigenic sin, an attribute of immune memory that leads to greater induction of antibodies specific to the first-encountered variant of an immunogen compared with subsequent variants. In this Review, recent findings on original antigenic sin are discussed in the context of SARS-CoV-2 evolution. Unanswered questions and future directions are highlighted, with an emphasis on the impact on disease outcome and vaccine design.
Collapse
|
13
|
Khoo WH, Jackson K, Phetsouphanh C, Zaunders JJ, Alquicira-Hernandez J, Yazar S, Ruiz-Diaz S, Singh M, Dhenni R, Kyaw W, Tea F, Merheb V, Lee FXZ, Burrell R, Howard-Jones A, Koirala A, Zhou L, Yuksel A, Catchpoole DR, Lai CL, Vitagliano TL, Rouet R, Christ D, Tang B, West NP, George S, Gerrard J, Croucher PI, Kelleher AD, Goodnow CG, Sprent JD, Powell JE, Brilot F, Nanan R, Hsu PS, Deenick EK, Britton PN, Phan TG. Tracking the clonal dynamics of SARS-CoV-2-specific T cells in children and adults with mild/asymptomatic COVID-19. Clin Immunol 2023; 246:109209. [PMID: 36539107 PMCID: PMC9758763 DOI: 10.1016/j.clim.2022.109209] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/28/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
Abstract
Children infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) develop less severe coronavirus disease 2019 (COVID-19) than adults. The mechanisms for the age-specific differences and the implications for infection-induced immunity are beginning to be uncovered. We show by longitudinal multimodal analysis that SARS-CoV-2 leaves a small footprint in the circulating T cell compartment in children with mild/asymptomatic COVID-19 compared to adult household contacts with the same disease severity who had more evidence of systemic T cell interferon activation, cytotoxicity and exhaustion. Children harbored diverse polyclonal SARS-CoV-2-specific naïve T cells whereas adults harbored clonally expanded SARS-CoV-2-specific memory T cells. A novel population of naïve interferon-activated T cells is expanded in acute COVID-19 and is recruited into the memory compartment during convalescence in adults but not children. This was associated with the development of robust CD4+ memory T cell responses in adults but not children. These data suggest that rapid clearance of SARS-CoV-2 in children may compromise their cellular immunity and ability to resist reinfection.
Collapse
Affiliation(s)
- Weng Hua Khoo
- Garvan Institute of Medical Research, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | | | | | - John J Zaunders
- Centre for Applied Medical Research, St Vincent's Hospital, Sydney, Australia
| | - José Alquicira-Hernandez
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, Australia; Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Seyhan Yazar
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, Australia
| | | | - Mandeep Singh
- Garvan Institute of Medical Research, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Rama Dhenni
- Garvan Institute of Medical Research, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Wunna Kyaw
- Garvan Institute of Medical Research, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Fiona Tea
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, Australia
| | - Vera Merheb
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, Australia
| | - Fiona X Z Lee
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, Australia
| | - Rebecca Burrell
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | | | - Archana Koirala
- Kids Research, The Children's Hospital at Westmead, Sydney, Australia
| | - Li Zhou
- Kids Research, The Children's Hospital at Westmead, Sydney, Australia
| | - Aysen Yuksel
- Kids Research, The Children's Hospital at Westmead, Sydney, Australia
| | - Daniel R Catchpoole
- Kids Research, The Children's Hospital at Westmead, Sydney, Australia; Discipline of Child and Adolescent Health, The University of Sydney, Sydney, Australia
| | - Catherine L Lai
- Kids Research, The Children's Hospital at Westmead, Sydney, Australia
| | | | - Romain Rouet
- Garvan Institute of Medical Research, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Daniel Christ
- Garvan Institute of Medical Research, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Benjamin Tang
- Department of Intensive Care Medicine, Nepean Hospital, Sydney, Australia; Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Sydney, Australia; Respiratory Tract Infection Research Node, Marie Bashir Institute for Infectious Diseases and Biosecurity, Sydney, Australia
| | - Nicholas P West
- Systems Biology and Data Science, Menzies Health Institute QLD, Griffith University, Parklands, Australia
| | - Shane George
- Departments of Emergency Medicine and Children's Critical Care, Gold Coast University Hospital, Southport, QLD, Australia; School of Medicine and Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - John Gerrard
- Department of Infectious Diseases and Immunology, Gold Coast University Hospital, Southport, QLD, Australia
| | - Peter I Croucher
- Garvan Institute of Medical Research, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | | | - Christopher G Goodnow
- Garvan Institute of Medical Research, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia; UNSW Cellular Genomics Futures Institute, UNSW Sydney, Sydney, Australia
| | - Jonathan D Sprent
- Garvan Institute of Medical Research, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Joseph E Powell
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, Australia; UNSW Cellular Genomics Futures Institute, UNSW Sydney, Sydney, Australia
| | - Fabienne Brilot
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, Australia; Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, Australia; Brain and Mind Centre, The University of Sydney, Sydney, Australia
| | - Ralph Nanan
- Charles Perkins Centre Nepean, University of Sydney, Sydney, Australia
| | - Peter S Hsu
- Kids Research, The Children's Hospital at Westmead, Sydney, Australia; Discipline of Child and Adolescent Health, The University of Sydney, Sydney, Australia
| | - Elissa K Deenick
- Garvan Institute of Medical Research, Sydney, Australia; Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Philip N Britton
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, Australia; The Children's Hospital at Westmead, Sydney Children's Hospitals Network, Sydney, Australia
| | - Tri Giang Phan
- Garvan Institute of Medical Research, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia.
| |
Collapse
|
14
|
McCarthy MW. Original antigen sin and COVID-19: implications for seasonal vaccination. Expert Opin Biol Ther 2022; 22:1353-1358. [PMID: 36243027 DOI: 10.1080/14712598.2022.2137402] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Original antigenic sin describes the phenomenon in which immunity against pathogens or antigens is shaped by the host's first exposure to a related pathogen or antigen. AREAS COVERED When primary immunity is boosted not by the homologous but by a cross-reacting vaccine, the newly formed antibodies may react better with the primary antigen than with the antigen actually eliciting the response. This form of immune imprinting, which has been observed with influenza, dengue, human immunodeficiency virus, and other pathogens, has profound implications for the approach to seasonal vaccination against a variety of diseases, including COVID-19. EXPERT OPINION Public health agencies and regulatory bodies have consistently recommended repeated vaccination every few months as a way to protect against COVID-19. However, the risks and benefits of this approach requires scrutiny given the concern for original antigenic sin in response to SARS-CoV-2. This manuscript examines what is known about immune imprinting and looks ahead to explore how this phenomenon may impact seasonal vaccination against emerging SARS-CoV-2 subvariants such as BA.4, BA.5, and BA.5.1, which have been associated increased transmissibility due to enhanced immune escape.
Collapse
|
15
|
Bigay J, Le Grand R, Martinon F, Maisonnasse P. Vaccine-associated enhanced disease in humans and animal models: Lessons and challenges for vaccine development. Front Microbiol 2022; 13:932408. [PMID: 36033843 PMCID: PMC9399815 DOI: 10.3389/fmicb.2022.932408] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
The fight against infectious diseases calls for the development of safe and effective vaccines that generate long-lasting protective immunity. In a few situations, vaccine-mediated immune responses may have led to exacerbated pathology upon subsequent infection with the pathogen targeted by the vaccine. Such vaccine-associated enhanced disease (VAED) has been reported, or at least suspected, in animal models, and in a few instances in humans, for vaccine candidates against the respiratory syncytial virus (RSV), measles virus (MV), dengue virus (DENV), HIV-1, simian immunodeficiency virus (SIV), feline immunodeficiency virus (FIV), severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1), and the Middle East respiratory syndrome coronavirus (MERS-CoV). Although alleviated by clinical and epidemiological evidence, a number of concerns were also initially raised concerning the short- and long-term safety of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is causing the ongoing COVID-19 pandemic. Although the mechanisms leading to this phenomenon are not yet completely understood, the individual and/or collective role of antibody-dependent enhancement (ADE), complement-dependent enhancement, and cell-dependent enhancement have been highlighted. Here, we review mechanisms that may be associated with the risk of VAED, which are important to take into consideration, both in the assessment of vaccine safety and in finding ways to define models and immunization strategies that can alleviate such concerns.
Collapse
Affiliation(s)
| | | | - Frédéric Martinon
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud-INSERM U1184, CEA, Fontenay-Aux-Roses, France
| | | |
Collapse
|
16
|
Dolton G, Rius C, Hasan MS, Wall A, Szomolay B, Behiry E, Whalley T, Southgate J, Fuller A, Morin T, Topley K, Tan LR, Goulder PJR, Spiller OB, Rizkallah PJ, Jones LC, Connor TR, Sewell AK. Emergence of immune escape at dominant SARS-CoV-2 killer T cell epitope. Cell 2022; 185:2936-2951.e19. [PMID: 35931021 PMCID: PMC9279490 DOI: 10.1016/j.cell.2022.07.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 05/08/2022] [Accepted: 07/07/2022] [Indexed: 01/06/2023]
Abstract
We studied the prevalent cytotoxic CD8 T cell response mounted against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Spike glycoprotein269-277 epitope (sequence YLQPRTFLL) via the most frequent human leukocyte antigen (HLA) class I worldwide, HLA A∗02. The Spike P272L mutation that has arisen in at least 112 different SARS-CoV-2 lineages to date, including in lineages classified as "variants of concern," was not recognized by the large CD8 T cell response seen across cohorts of HLA A∗02+ convalescent patients and individuals vaccinated against SARS-CoV-2, despite these responses comprising of over 175 different individual T cell receptors. Viral escape at prevalent T cell epitopes restricted by high frequency HLAs may be particularly problematic when vaccine immunity is focused on a single protein such as SARS-CoV-2 Spike, providing a strong argument for inclusion of multiple viral proteins in next generation vaccines and highlighting the need for monitoring T cell escape in new SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Garry Dolton
- Division of Infection and Immunity, Cardiff University School of Medicine, CF14 4XN Cardiff, Wales, UK
| | - Cristina Rius
- Division of Infection and Immunity, Cardiff University School of Medicine, CF14 4XN Cardiff, Wales, UK
| | - Md Samiul Hasan
- Division of Infection and Immunity, Cardiff University School of Medicine, CF14 4XN Cardiff, Wales, UK
| | - Aaron Wall
- Division of Infection and Immunity, Cardiff University School of Medicine, CF14 4XN Cardiff, Wales, UK
| | - Barbara Szomolay
- Division of Infection and Immunity, Cardiff University School of Medicine, CF14 4XN Cardiff, Wales, UK; Systems Immunology Research Institute, Cardiff University, CF14 4XN Cardiff, Wales, UK
| | - Enas Behiry
- Division of Infection and Immunity, Cardiff University School of Medicine, CF14 4XN Cardiff, Wales, UK
| | - Thomas Whalley
- School of Biosciences, Cardiff University, CF10 3AX Cardiff, Wales, UK
| | - Joel Southgate
- School of Biosciences, Cardiff University, CF10 3AX Cardiff, Wales, UK
| | - Anna Fuller
- Division of Infection and Immunity, Cardiff University School of Medicine, CF14 4XN Cardiff, Wales, UK
| | - Théo Morin
- Division of Infection and Immunity, Cardiff University School of Medicine, CF14 4XN Cardiff, Wales, UK
| | - Katie Topley
- Division of Infection and Immunity, Cardiff University School of Medicine, CF14 4XN Cardiff, Wales, UK
| | - Li Rong Tan
- Division of Infection and Immunity, Cardiff University School of Medicine, CF14 4XN Cardiff, Wales, UK
| | - Philip J R Goulder
- Department of Paediatrics, University of Oxford, OX3 9DU Oxford, England, UK
| | - Owen B Spiller
- Division of Infection and Immunity, Cardiff University School of Medicine, CF14 4XN Cardiff, Wales, UK
| | - Pierre J Rizkallah
- Division of Infection and Immunity, Cardiff University School of Medicine, CF14 4XN Cardiff, Wales, UK
| | - Lucy C Jones
- Division of Infection and Immunity, Cardiff University School of Medicine, CF14 4XN Cardiff, Wales, UK; Centre for Clinical Research, Royal Glamorgan Hospital, Ynysmaerdy CF72 8XR, UK
| | - Thomas R Connor
- Systems Immunology Research Institute, Cardiff University, CF14 4XN Cardiff, Wales, UK; School of Biosciences, Cardiff University, CF10 3AX Cardiff, Wales, UK; Pathogen genomics Unit, Public Health Wales NHS Trust, CF14 4XW Cardiff, Wales, UK.
| | - Andrew K Sewell
- Division of Infection and Immunity, Cardiff University School of Medicine, CF14 4XN Cardiff, Wales, UK; Systems Immunology Research Institute, Cardiff University, CF14 4XN Cardiff, Wales, UK.
| |
Collapse
|
17
|
Lv H, So RTY, Teo QW, Yuan M, Liu H, Lee CCD, Yip GK, Ng WW, Wilson IA, Peiris M, Wu NC, Mok CKP. Neutralizing Antibody Response to Sarbecovirus Is Delayed in Sequential Heterologous Immunization. Viruses 2022; 14:1382. [PMID: 35891363 PMCID: PMC9318566 DOI: 10.3390/v14071382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 02/01/2023] Open
Abstract
Antigenic imprinting, which describes the bias of the antibody response due to previous immune history, can influence vaccine effectiveness. While this phenomenon has been reported for viruses such as influenza, there is little understanding of how prior immune history affects the antibody response to SARS-CoV-2. This study provides evidence for antigenic imprinting through immunization with two Sarbecoviruses, the subgenus that includes SARS-CoV-2. Mice were immunized subsequently with two antigenically distinct Sarbecovirus strains, namely SARS-CoV-1 and SARS-CoV-2. We found that sequential heterologous immunization induced cross-reactive binding antibodies for both viruses and delayed the emergence of neutralizing antibody responses against the booster strain. Our results provide fundamental knowledge about the immune response to Sarbecovirus and important insights into the development of pan-sarbecovirus vaccines and guiding therapeutic interventions.
Collapse
Affiliation(s)
- Huibin Lv
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (H.L.); (R.T.Y.S.); (Q.W.T.); (G.K.Y.); (W.W.N.); (M.P.)
| | - Ray T. Y. So
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (H.L.); (R.T.Y.S.); (Q.W.T.); (G.K.Y.); (W.W.N.); (M.P.)
| | - Qi Wen Teo
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (H.L.); (R.T.Y.S.); (Q.W.T.); (G.K.Y.); (W.W.N.); (M.P.)
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Meng Yuan
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA; (M.Y.); (H.L.); (C.-C.D.L.); (I.A.W.)
| | - Hejun Liu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA; (M.Y.); (H.L.); (C.-C.D.L.); (I.A.W.)
| | - Chang-Chun D. Lee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA; (M.Y.); (H.L.); (C.-C.D.L.); (I.A.W.)
| | - Garrick K. Yip
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (H.L.); (R.T.Y.S.); (Q.W.T.); (G.K.Y.); (W.W.N.); (M.P.)
| | - Wilson W. Ng
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (H.L.); (R.T.Y.S.); (Q.W.T.); (G.K.Y.); (W.W.N.); (M.P.)
| | - Ian A. Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA; (M.Y.); (H.L.); (C.-C.D.L.); (I.A.W.)
- The Skaggs Institute for Chemical Biology, the Scripps Research Institute, La Jolla, CA 92037, USA
| | - Malik Peiris
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (H.L.); (R.T.Y.S.); (Q.W.T.); (G.K.Y.); (W.W.N.); (M.P.)
| | - Nicholas C. Wu
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Center for Biophysics and Computational Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Chris Ka Pun Mok
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
18
|
Luria-Pérez R, Sánchez-Vargas LA, Muñoz-López P, Mellado-Sánchez G. Mucosal Vaccination: A Promising Alternative Against Flaviviruses. Front Cell Infect Microbiol 2022; 12:887729. [PMID: 35782117 PMCID: PMC9241634 DOI: 10.3389/fcimb.2022.887729] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/10/2022] [Indexed: 12/15/2022] Open
Abstract
The Flaviviridae are a family of positive-sense, single-stranded RNA enveloped viruses, and their members belong to a single genus, Flavivirus. Flaviviruses are found in mosquitoes and ticks; they are etiological agents of: dengue fever, Japanese encephalitis, West Nile virus infection, Zika virus infection, tick-borne encephalitis, and yellow fever, among others. Only a few flavivirus vaccines have been licensed for use in humans: yellow fever, dengue fever, Japanese encephalitis, tick-borne encephalitis, and Kyasanur forest disease. However, improvement is necessary in vaccination strategies and in understanding of the immunological mechanisms involved either in the infection or after vaccination. This is especially important in dengue, due to the immunological complexity of its four serotypes, cross-reactive responses, antibody-dependent enhancement, and immunological interference. In this context, mucosal vaccines represent a promising alternative against flaviviruses. Mucosal vaccination has several advantages, as inducing long-term protective immunity in both mucosal and parenteral tissues. It constitutes a friendly route of antigen administration because it is needle-free and allows for a variety of antigen delivery systems. This has promoted the development of several ways to stimulate immunity through the direct administration of antigens (e.g., inactivated virus, attenuated virus, subunits, and DNA), non-replicating vectors (e.g., nanoparticles, liposomes, bacterial ghosts, and defective-replication viral vectors), and replicating vectors (e.g., Salmonella enterica, Lactococcus lactis, Saccharomyces cerevisiae, and viral vectors). Because of these characteristics, mucosal vaccination has been explored for immunoprophylaxis against pathogens that enter the host through mucosae or parenteral areas. It is suitable against flaviviruses because this type of immunization can stimulate the parenteral responses required after bites from flavivirus-infected insects. This review focuses on the advantages of mucosal vaccine candidates against the most relevant flaviviruses in either humans or animals, providing supporting data on the feasibility of this administration route for future clinical trials.
Collapse
Affiliation(s)
- Rosendo Luria-Pérez
- Hospital Infantil de México Federico Gómez, Unidad de Investigación en Enfermedades Hemato-Oncológicas, Ciudad de México, Mexico
| | - Luis A. Sánchez-Vargas
- Department of Cell and Molecular Biology, Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, United States
| | - Paola Muñoz-López
- Hospital Infantil de México Federico Gómez, Unidad de Investigación en Enfermedades Hemato-Oncológicas, Ciudad de México, Mexico
- Posgrado en Biomedicina y Biotecnología Molecular, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Gabriela Mellado-Sánchez
- Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
- Laboratorio Nacional para Servicios Especializados de Investigación, Desarrollo e Innovación (I+D+i) para Farmoquímicos y Biotecnológicos, LANSEIDI-FarBiotec-CONACyT, Ciudad de México, Mexico
| |
Collapse
|
19
|
Zamora-Ceballos M, Moreno N, Gil-Cantero D, Castón JR, Blanco E, Bárcena J. Immunogenicity of Multi-Target Chimeric RHDV Virus-like Particles Delivering Foreign B-Cell Epitopes. Vaccines (Basel) 2022; 10:vaccines10020229. [PMID: 35214688 PMCID: PMC8875457 DOI: 10.3390/vaccines10020229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/29/2022] [Accepted: 01/31/2022] [Indexed: 11/16/2022] Open
Abstract
The rabbit hemorrhagic disease virus (RHDV) vaccine platform is a nanoparticle composed of 180 copies of the viral capsid protein, VP60, self-assembled into virus-like particles (VLPs). RHDV VLPs are able to accept the simultaneous incorporation of target epitopes at different insertion sites. The resulting chimeric RHDV VLPs displaying immunogenic foreign antigens have been shown to induce specific protective immune responses against inserted heterologous T-cytotoxic and B-cell epitopes in the mouse and pig models. In this study, we explored whether RHDV-based engineered VLPs can be developed as efficient multivalent vaccines co-delivering different foreign B-cell antigens. We generated bivalent chimeric RHDV VLPs displaying two model B-cell epitopes at different surface-exposed insertion sites, as well as the corresponding monovalent chimeric VLPs. The immunogenic potential of the bivalent chimeric VLPs versus the monovalent constructs was assessed in the mouse model. We found that the bivalent chimeric VLPs elicited a strong and balanced antibody response towards the two target epitopes tested, although slight reductions were observed in the levels of specific serum antibody titers induced by bivalent chimeric VLPs as compared with the corresponding monovalent constructs. These results suggest that RHDV VLPs could represent a promising platform for the development of efficient multivalent vaccines.
Collapse
Affiliation(s)
- María Zamora-Ceballos
- Instituto Centro de Investigación en Sanidad Animal (CISA-INIA/CSIC), Valdeolmos, 28130 Madrid, Spain; (M.Z.-C.); (N.M.); (E.B.)
| | - Noelia Moreno
- Instituto Centro de Investigación en Sanidad Animal (CISA-INIA/CSIC), Valdeolmos, 28130 Madrid, Spain; (M.Z.-C.); (N.M.); (E.B.)
| | - David Gil-Cantero
- Department of Structure of Macromolecules, Centro Nacional de Biotecnología/CSIC, Cantoblanco, 28049 Madrid, Spain; (D.G.-C.); (J.R.C.)
| | - José R. Castón
- Department of Structure of Macromolecules, Centro Nacional de Biotecnología/CSIC, Cantoblanco, 28049 Madrid, Spain; (D.G.-C.); (J.R.C.)
| | - Esther Blanco
- Instituto Centro de Investigación en Sanidad Animal (CISA-INIA/CSIC), Valdeolmos, 28130 Madrid, Spain; (M.Z.-C.); (N.M.); (E.B.)
| | - Juan Bárcena
- Instituto Centro de Investigación en Sanidad Animal (CISA-INIA/CSIC), Valdeolmos, 28130 Madrid, Spain; (M.Z.-C.); (N.M.); (E.B.)
- Correspondence: ; Tel.: +34-916-202-300
| |
Collapse
|
20
|
Gartlan C, Tipton T, Salguero FJ, Sattentau Q, Gorringe A, Carroll MW. Vaccine-Associated Enhanced Disease and Pathogenic Human Coronaviruses. Front Immunol 2022; 13:882972. [PMID: 35444667 PMCID: PMC9014240 DOI: 10.3389/fimmu.2022.882972] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/14/2022] [Indexed: 01/14/2023] Open
Abstract
Vaccine-associated enhanced disease (VAED) is a difficult phenomenon to define and can be confused with vaccine failure. Using studies on respiratory syncytial virus (RSV) vaccination and dengue virus infection, we highlight known and theoretical mechanisms of VAED, including antibody-dependent enhancement (ADE), antibody-enhanced disease (AED) and Th2-mediated pathology. We also critically review the literature surrounding this phenomenon in pathogenic human coronaviruses, including MERS-CoV, SARS-CoV-1 and SARS-CoV-2. Poor quality histopathological data and a lack of consistency in defining severe pathology and VAED in preclinical studies of MERS-CoV and SARS-CoV-1 vaccines in particular make it difficult to interrogate potential cases of VAED. Fortuitously, there have been only few reports of mild VAED in SARS-CoV-2 vaccination in preclinical models and no observations in their clinical use. We describe the problem areas and discuss methods to improve the characterisation of VAED in the future.
Collapse
Affiliation(s)
- Cillian Gartlan
- Wellcome Centre for Human Genetics and Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Tom Tipton
- Wellcome Centre for Human Genetics and Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Francisco J Salguero
- Research and Evaluation, UK Health Security Agency, Porton Down, Salisbury, United Kingdom
| | - Quentin Sattentau
- The Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Andrew Gorringe
- Research and Evaluation, UK Health Security Agency, Porton Down, Salisbury, United Kingdom
| | - Miles W Carroll
- Wellcome Centre for Human Genetics and Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
21
|
Chen H, Smith M, Herz J, Li T, Hasley R, Le Saout C, Zhu Z, Cheng J, Gronda A, Martina JA, Irusta PM, Karpova T, McGavern DB, Catalfamo M. The role of protease-activated receptor 1 signaling in CD8 T cell effector functions. iScience 2021; 24:103387. [PMID: 34841225 PMCID: PMC8605340 DOI: 10.1016/j.isci.2021.103387] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 09/28/2021] [Accepted: 10/28/2021] [Indexed: 12/15/2022] Open
Abstract
CD8 T cells are essential for adaptive immunity against viral infections. Protease activated receptor 1 (PAR1) is expressed by CD8 T cells; however, its role in T cell effector function is not well defined. Here we show that in human CD8 T cells, PAR1 stimulation accelerates calcium mobilization. Furthermore, PAR1 is involved in cytotoxic T cell function by facilitating granule trafficking via actin polymerization and repositioning of the microtubule organizing center (MTOC) toward the immunological synapse. In vivo, PAR1-/- mice have reduced cytokine-producing T cells in response to a lymphocytic choriomeningitis virus (LCMV) infection and fail to efficiently control the virus. Specific deletion of PAR1 in LCMV GP33-specific CD8 T cells results in reduced expansion and diminished effector function. These data demonstrate that PAR1 plays a role in T cell activation and function, and this pathway could represent a new therapeutic strategy to modulate CD8 T cell effector function.
Collapse
Affiliation(s)
- Hui Chen
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, USA
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mindy Smith
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jasmin Herz
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Tong Li
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, USA
| | - Rebecca Hasley
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Cecile Le Saout
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ziang Zhu
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, USA
| | - Jie Cheng
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, USA
| | - Andres Gronda
- Department of Human Science, Georgetown University, Washington, DC, USA
| | - José A. Martina
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Pablo M. Irusta
- Department of Human Science, Georgetown University, Washington, DC, USA
| | - Tatiana Karpova
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dorian B. McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Marta Catalfamo
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, USA
| |
Collapse
|
22
|
Abstract
Exceptional efforts have been undertaken to shed light into the biology of adaptive immune responses to SARS-CoV-2. T cells occupy a central role in adaptive immunity to mediate helper functions to different arms of the immune system and are fundamental to mediate protection, control, and clearance of most viral infections. Even though many questions remain unsolved, there is a growing literature linking specific T cell characteristics to differential COVID-19 severity and vaccine outcome. In this review, we summarize our current understanding of CD4+ and CD8+ T cell responses in acute and convalescent COVID-19. Further, we discuss the T cell literature coupled to pre-existing immunity and vaccines and highlight the need to look beyond blood to fully understand how T cells function in the tissue space.
Collapse
Affiliation(s)
- Julia Niessl
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Takuya Sekine
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Marcus Buggert
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
23
|
Neidleman J, Luo X, McGregor M, Xie G, Murray V, Greene WC, Lee SA, Roan NR. mRNA vaccine-induced T cells respond identically to SARS-CoV-2 variants of concern but differ in longevity and homing properties depending on prior infection status. eLife 2021; 10:e72619. [PMID: 34636722 PMCID: PMC8545397 DOI: 10.7554/elife.72619] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/05/2021] [Indexed: 12/11/2022] Open
Abstract
While mRNA vaccines are proving highly efficacious against SARS-CoV-2, it is important to determine how booster doses and prior infection influence the immune defense they elicit, and whether they protect against variants. Focusing on the T cell response, we conducted a longitudinal study of infection-naïve and COVID-19 convalescent donors before vaccination and after their first and second vaccine doses, using a high-parameter CyTOF analysis to phenotype their SARS-CoV-2-specific T cells. Vaccine-elicited spike-specific T cells responded similarly to stimulation by spike epitopes from the ancestral, B.1.1.7 and B.1.351 variant strains, both in terms of cell numbers and phenotypes. In infection-naïve individuals, the second dose boosted the quantity and altered the phenotypic properties of SARS-CoV-2-specific T cells, while in convalescents the second dose changed neither. Spike-specific T cells from convalescent vaccinees differed strikingly from those of infection-naïve vaccinees, with phenotypic features suggesting superior long-term persistence and ability to home to the respiratory tract including the nasopharynx. These results provide reassurance that vaccine-elicited T cells respond robustly to emerging viral variants, confirm that convalescents may not need a second vaccine dose, and suggest that vaccinated convalescents may have more persistent nasopharynx-homing SARS-CoV-2-specific T cells compared to their infection-naïve counterparts.
Collapse
Affiliation(s)
- Jason Neidleman
- Department or Urology, University of California, San FranciscoSan FranciscoUnited States
- Gladstone Institute of VirologySan FranciscoUnited States
| | - Xiaoyu Luo
- Gladstone Institute of VirologySan FranciscoUnited States
| | - Matthew McGregor
- Department or Urology, University of California, San FranciscoSan FranciscoUnited States
- Gladstone Institute of VirologySan FranciscoUnited States
| | - Guorui Xie
- Department or Urology, University of California, San FranciscoSan FranciscoUnited States
- Gladstone Institute of VirologySan FranciscoUnited States
| | - Victoria Murray
- University of California, San FranciscoSan FranciscoUnited States
| | | | - Sulggi A Lee
- Medicine, University of California, San FranciscoSan FranciscoUnited States
| | - Nadia R Roan
- Department or Urology, University of California, San FranciscoSan FranciscoUnited States
- Gladstone Institute of VirologySan FranciscoUnited States
| |
Collapse
|
24
|
Abstract
CoVID-19 is a multi-symptomatic disease which has made a global impact due to its ability to spread rapidly, and its relatively high mortality rate. Beyond the heroic efforts to develop vaccines, which we do not discuss herein, the response of scientists and clinicians to this complex problem has reflected the need to detect CoVID-19 rapidly, to diagnose patients likely to show adverse symptoms, and to treat severe and critical CoVID-19. Here we aim to encapsulate these varied and sometimes conflicting approaches and the resulting data in terms of chemistry and biology. In the process we highlight emerging concepts, and potential future applications that may arise out of this immense effort.
Collapse
Affiliation(s)
| | - Yimon Aye
- Swiss Federal Institute of Technology in Lausanne (EPFL)1015LausanneSwitzerland
| |
Collapse
|
25
|
Neidleman J, Luo X, McGregor M, Xie G, Murray V, Greene WC, Lee SA, Roan NR. mRNA vaccine-induced T cells respond identically to SARS-CoV-2 variants of concern but differ in longevity and homing properties depending on prior infection status. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.05.12.443888. [PMID: 34013277 PMCID: PMC8132283 DOI: 10.1101/2021.05.12.443888] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
While mRNA vaccines are proving highly efficacious against SARS-CoV-2, it is important to determine how booster doses and prior infection influence the immune defense they elicit, and whether they protect against variants. Focusing on the T cell response, we conducted a longitudinal study of infection-naïve and COVID-19 convalescent donors before vaccination and after their first and second vaccine doses, using a high-parameter CyTOF analysis to phenotype their SARS-CoV-2-specific T cells. Vaccine-elicited spike-specific T cells responded similarly to stimulation by spike epitopes from the ancestral, B.1.1.7 and B.1.351 variant strains, both in terms of cell numbers and phenotypes. In infection-naïve individuals, the second dose boosted the quantity and altered the phenotypic properties of SARS-CoV-2-specific T cells, while in convalescents the second dose changed neither. Spike-specific T cells from convalescent vaccinees differed strikingly from those of infection-naïve vaccinees, with phenotypic features suggesting superior long-term persistence and ability to home to the respiratory tract including the nasopharynx. These results provide reassurance that vaccine-elicited T cells respond robustly to emerging viral variants, confirm that convalescents may not need a second vaccine dose, and suggest that vaccinated convalescents may have more persistent nasopharynx-homing SARS-CoV-2-specific T cells compared to their infection-naïve counterparts.
Collapse
Affiliation(s)
- Jason Neidleman
- Gladstone Institute of Virology, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, CA, USA
| | - Xiaoyu Luo
- Gladstone Institute of Virology, San Francisco, CA, USA
| | - Matthew McGregor
- Gladstone Institute of Virology, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, CA, USA
| | - Guorui Xie
- Gladstone Institute of Virology, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, CA, USA
| | - Victoria Murray
- Zuckerberg San Francisco General Hospital and the University of California, San Francisco, CA, USA
| | - Warner C. Greene
- Gladstone Institute of Virology, San Francisco, CA, USA
- Departments of Medicine, and Microbiology and Immunology, University of California, San Francisco, CA, USA
| | | | - Nadia R. Roan
- Gladstone Institute of Virology, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, CA, USA
| |
Collapse
|
26
|
Neoantigen Cancer Vaccines: Real Opportunity or Another Illusion? Arch Immunol Ther Exp (Warsz) 2021; 69:12. [PMID: 33909124 PMCID: PMC8080209 DOI: 10.1007/s00005-021-00615-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/17/2021] [Indexed: 02/07/2023]
Abstract
In this communication, we will analyze some important factors and immunological phenomena related to neoantigen cancer vaccines, with particular emphasis on recently published Phase I clinical trials. Several obstacles and issues are addressed that challenge the current paradigm and inquire if neoantigens, which are essentially single-use vaccine candidates, are legitimate targets to induce protective immune responses with regard to the evolving mutational landscape. We also share insights into the striking similarities between cancer and antigenically variable pathogens and suggest that any successful vaccine against either should demonstrate a similar property: efficient induction of a diverse pool of immune cells equipped to prevent immune escape. Hence, to confront antigenic variability directly, we have employed our innovative vaccine concept, Variable Epitope Libraries, composed of large combinatorial libraries of heavily mutated epitopes, as a "universal" vaccine platform. Collectively, we offer critical analyses on key issues, which ultimately reflect on the prospective clinical relevance of personalized neoantigen vaccines which is still undefined.
Collapse
|
27
|
Rha MS, Kim AR, Shin EC. SARS-CoV-2-Specific T Cell Responses in Patients with COVID-19 and Unexposed Individuals. Immune Netw 2021; 21:e2. [PMID: 33728095 PMCID: PMC7937509 DOI: 10.4110/in.2021.21.e2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/05/2021] [Accepted: 02/07/2021] [Indexed: 02/06/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection causes coronavirus disease 2019 (COVID-19), an ongoing pandemic disease. In the current review, we describe SARS-CoV-2-specific CD4+ and CD8+ T-cell responses in acute and convalescent COVID-19 patients. We also discuss the relationships between COVID-19 severity and SARS-CoV-2-specific T-cell responses and summarize recent reports regarding SARS-CoV-2-reactive T cells in SARS-CoV-2-unexposed individuals. These T cells may be cross-reactive cells primed by previous infection with human common-cold coronaviruses. Finally, we outline SARS-CoV-2-specific T-cell responses in the context of vaccination. A better understanding of SARS-CoV-2-specific T-cell responses is needed to develop effective vaccines and therapeutics.
Collapse
Affiliation(s)
- Min-Seok Rha
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - A Reum Kim
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Eui-Cheol Shin
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea.,The Center for Epidemic Preparedness, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| |
Collapse
|
28
|
Bhatt P, Sabeena SP, Varma M, Arunkumar G. Current Understanding of the Pathogenesis of Dengue Virus Infection. Curr Microbiol 2021; 78:17-32. [PMID: 33231723 PMCID: PMC7815537 DOI: 10.1007/s00284-020-02284-w] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 11/04/2020] [Indexed: 12/26/2022]
Abstract
The pathogenesis of dengue virus infection is attributed to complex interplay between virus, host genes and host immune response. Host factors such as antibody-dependent enhancement (ADE), memory cross-reactive T cells, anti-DENV NS1 antibodies, autoimmunity as well as genetic factors are major determinants of disease susceptibility. NS1 protein and anti-DENV NS1 antibodies were believed to be responsible for pathogenesis of severe dengue. The cytokine response of cross-reactive CD4+ T cells might be altered by the sequential infection with different DENV serotypes, leading to further elevation of pro-inflammatory cytokines contributing a detrimental immune response. Fcγ receptor-mediated antibody-dependent enhancement (ADE) results in release of cytokines from immune cells leading to vascular endothelial cell dysfunction and increased vascular permeability. Genomic variation of dengue virus and subgenomic flavivirus RNA (sfRNA) suppressing host immune response are viral determinants of disease severity. Dengue infection can lead to the generation of autoantibodies against DENV NS1antigen, DENV prM, and E proteins, which can cross-react with several self-antigens such as plasminogen, integrin, and platelet cells. Apart from viral factors, several host genetic factors and gene polymorphisms also have a role to play in pathogenesis of DENV infection. This review article highlights the various factors responsible for the pathogenesis of dengue and also highlights the recent advances in the field related to biomarkers which can be used in future for predicting severe disease outcome.
Collapse
Affiliation(s)
- Puneet Bhatt
- Manipal Institute of Virology, Manipal Academy of Higher Education, Manipal, Karnataka 576104 India
| | | | - Muralidhar Varma
- Dept of Infectious Diseases, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka 576101 India
| | - Govindakarnavar Arunkumar
- Manipal Institute of Virology, Manipal Academy of Higher Education, Manipal, Karnataka 576104 India
- Present Address: WHO Country Office, Kathmandu, Nepal
| |
Collapse
|
29
|
Khan BA, Saifullah, Lail A, Khan S. Sub-genomic analysis of Chikungunya virus E2 mutations in Pakistani isolates potentially modulating B-cell & T-Cell immune response. Pak J Med Sci 2020; 37:93-98. [PMID: 33437257 PMCID: PMC7794161 DOI: 10.12669/pjms.37.1.3236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background & Objectives: The Chikungunya virus (CHIKV) transmitted to the humans through Aedes species of the mosquitoes. In December 2016, a severe outbreak reported from Pakistan. However, there is no vaccine or anti-viral treatment currently available so host immune response against CHIKV gained significant interest. Therefore, this study was conducted to identify the mutations in CHIKV E2 region of currently circulating Pakistani strains & determine their potential immunogenicity in Pakistani population. Methods: It was a cross sectional study in which a total of 60 CHIKV PCR positive samples were collected from Molecular Department of Pathology, Dow University of Health Sciences (DUHS), Karachi during November 2017 to February 2018. CHIKV E2 gene was amplified by PCR & sequenced. Sequences were analyzed by using bioinformatic tools followed by epitope prediction in E2 sequences by In-silico immunoinformatic approach. Results: Several single nucleotide variations (SNVs) were identified in Pakistani isolates with six novel mutations in E2 sequences. Immunoinformatic analyses showed more proteasomal sites, CTL & B-Cell epitopes in Pakistani strains with respect to S27 prototype with 69.4% population coverage against these epitopes in Pakistan. The study also identified key mutations responsible for generation of unique epitopes and HLA restriction in Pakistani isolates. The strain specific mutations revealed the current outbreak was caused by ESCA.IOL lineage of CHIKV. Conclusion: The evolution of E2 protein in Pakistani strains has increased its immunogenicity in comparison to ancestral s27 strain. The identification of most immunogenic and conserved epitopes with high population coverage has high potential to be used in vaccine development against these local strains.
Collapse
Affiliation(s)
- Bilal Ahmed Khan
- Bilal Ahmed Khan, M.Phil. Department of Biotechnology, University of Karachi, Karachi, Pakistan, Department of Pathology, Dow University of Health Sciences, Karachi, Pakistan
| | - Saifullah
- Dr. Saifullah, Ph.D. Department of Biotechnology, University of Karachi, Karachi, Pakistan
| | - Amanullah Lail
- Dr. Amanullah Lail, FCPS. Department of Pediatrics, Dow University of Health Sciences, Karachi, Pakistan
| | - Saeed Khan
- Prof. Dr. Saeed Khan, Ph.D. Department of Pathology, Dow University of Health Sciences, Karachi, Pakistan
| |
Collapse
|
30
|
Karlsson AC, Humbert M, Buggert M. The known unknowns of T cell immunity to COVID-19. Sci Immunol 2020; 5:5/53/eabe8063. [PMID: 33208380 DOI: 10.1126/sciimmunol.abe8063] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 11/13/2020] [Indexed: 12/13/2022]
Abstract
Tremendous progress has been made in understanding the role of T cell immunity in acute and convalescent COVID-19 infection. Here we shed light on the "known unknowns" of pre-existing and acquired T cell responses in relation to acute and convalescent SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Annika C Karlsson
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Marion Humbert
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Marcus Buggert
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
31
|
Naidu SAG, Clemens RA, Pressman P, Zaigham M, Kadkhoda K, Davies KJA, Naidu AS. COVID-19 during Pregnancy and Postpartum. J Diet Suppl 2020; 19:115-142. [PMID: 33164601 DOI: 10.1080/19390211.2020.1834049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Coronavirus Disease 2019 (COVID-19) triggered by Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) infection has been declared a pandemic by the World Health Organization (WHO) on March 11, 2020. Oxidative stress and its related metabolic syndromes are potential risk factors in the susceptibility to, and severity of COVID-19. In concert with the earliest reports of COVID-19, obstetricians started to diagnose and treat SARS-CoV-2 infections during pregnancy ("COVID-19-Pregnancy"). High metabolic demand to sustain normal fetal development increases the burden of oxidative stress in pregnancy. Intracellular redox changes intertwined with acute phase responses at the maternal-fetal interface could amplify during pregnancy. Interestingly, mother-to-fetus transmission of SARS-CoV-2 has not been detected in most of the COVID-19-Pregnancy cases. This relative absence of vertical transmission may be related to the presence of lactoferrin in the placenta, amniotic fluid, and lacteal secretions. However, the cytokine-storm induced during COVID-19-Pregnancy may cause severe inflammatory damage to the fetus, and if uncontrolled, may later result in autism spectrum-like disorders and brain development abnormalities in neonates. Considering this serious health threat to child growth and development, the prevention of COVID-19 during pregnancy should be considered a high priority. This review summarizes the intricate virulence factors of COVID-19 and elucidate its pathobiological spectrum during pregnancy and postpartum periods with a focus on the putative and complex roles of endogenous and exogenous lactoferrin in conferring immunological advantage to the host.
Collapse
Affiliation(s)
| | - Roger A Clemens
- University of Southern California, School of Pharmacy, Los Angeles, CA, USA
| | | | - Mehreen Zaigham
- Department of Obstetrics & Gynecology, Skåne University Hospital, Malmö, Sweden
| | - Kamran Kadkhoda
- Immunopathology Laboratory, Robert J. Tomisch Pathology & Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Kelvin J A Davies
- Division of Biogerontology, Leonard Davis School of Gerontology, The University of Southern California, Los Angeles, CA, USA.,Division of Molecular & Computational Biology, Dornsife College of Letters, Arts, and Sciences, The University of Southern California, Los Angeles, CA, USA.,Department Biochemistry & Molecular Medicine, Keck School of Medicine of USC, The University of Southern California, Los Angeles, CA, USA
| | | |
Collapse
|
32
|
Roncati L, Palmieri B. What about the original antigenic sin of the humans versus SARS-CoV-2? Med Hypotheses 2020; 142:109824. [PMID: 32408068 PMCID: PMC7204740 DOI: 10.1016/j.mehy.2020.109824] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 04/17/2020] [Accepted: 05/06/2020] [Indexed: 01/12/2023]
Affiliation(s)
- Luca Roncati
- Department of Pathology and Surgery, University Hospital of Modena, Modena, Italy.
| | - Beniamino Palmieri
- Department of Pathology and Surgery, University Hospital of Modena, Modena, Italy
| |
Collapse
|
33
|
Morsy S, Hashan MR, Hieu TH, Mohammed AT, Elawady SS, Ghosh P, Elgendy MA, Le HH, Hamad WMA, Iqtadar S, Dumre SP, Hirayama K, Huy NT. The association between dengue viremia kinetics and dengue severity: A systemic review and meta-analysis. Rev Med Virol 2020; 30:1-10. [PMID: 32856357 DOI: 10.1002/rmv.2121] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 12/29/2022]
Abstract
In this study, we aim to assess the association of dengue viremia with dengue severity. The study protocol was developed and registered in PROSPERO (CRD42016039864). We searched nine databases to find potential papers. Studies meeting the inclusion criteria were included. We, based our analysis on three outcomes which are disease severity, dengue serotype and disease infection type. Thirty studies with 3316 patients were included. Our analysis revealed that viremia is significantly higher in dengue hemorrhagic fever patients than dengue fever in days 5 to 6. Regarding the serotype of dengue, the maximum viremia titre of serotype 1 was significantly higher than serotype 3 and the viremia in dengue serotype 2 was significantly higher than serotype 4 in days 2 to 4. However, comparison of the daily viremia level between the primary and secondary dengue infection revealed that secondary infection was significantly higher than the primary infection on seventh day and on the eighth day. Viremia is strongly associated with disease severity and type of infection which gave viremia a high indicative power to be used as a clinical predictor. Dengue serotype is also associated with viral load with higher viremia in DENV-2/1.
Collapse
Affiliation(s)
- Sara Morsy
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Tanta University, Tanta, Egypt.,Online research Club (http://www.onlineresearchclub.org), Nagasaki, Japan
| | - Mohammad Rashidul Hashan
- Online research Club (http://www.onlineresearchclub.org), Nagasaki, Japan.,Respiratory and Enteric Infections Department, Infectious Disease Division, International Centre for Diarrheal Disease Research, Dhaka, Bangladesh
| | - Truong Hong Hieu
- Online research Club (http://www.onlineresearchclub.org), Nagasaki, Japan.,University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Abdelrahman Tarek Mohammed
- Online research Club (http://www.onlineresearchclub.org), Nagasaki, Japan.,Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Sameh Samir Elawady
- Online research Club (http://www.onlineresearchclub.org), Nagasaki, Japan.,Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Prithwish Ghosh
- Online research Club (http://www.onlineresearchclub.org), Nagasaki, Japan.,College Medicine and Sagore Dutta Hospital, West Bengal University of Health Science, West Bengal, India
| | - Manal A Elgendy
- Online research Club (http://www.onlineresearchclub.org), Nagasaki, Japan.,Biochemistry and Molecular Biology, Ain Shams University, Cairo, Egypt
| | - Huu-Hoai Le
- Online research Club (http://www.onlineresearchclub.org), Nagasaki, Japan.,Saigon General Hospital, Ho Chi Minh City, Vietnam
| | - Walid Mohamed Attiah Hamad
- Online research Club (http://www.onlineresearchclub.org), Nagasaki, Japan.,Internal Medicine Department Infectious Diseases Unit, Zagazig University, Zagazig, Egypt
| | - Somia Iqtadar
- Online research Club (http://www.onlineresearchclub.org), Nagasaki, Japan.,Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Shyam Prakash Dumre
- Department of Immunogenetics, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Kenji Hirayama
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Leading Graduate School Program, and Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Nguyen Tien Huy
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Leading Graduate School Program, and Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.,Institute of Research and Development, Duy Tan University, Da Nang, Vietnam
| |
Collapse
|
34
|
Nguyen A, David JK, Maden SK, Wood MA, Weeder BR, Nellore A, Thompson RF. Human Leukocyte Antigen Susceptibility Map for Severe Acute Respiratory Syndrome Coronavirus 2. J Virol 2020; 94:e00510-20. [PMID: 32303592 PMCID: PMC7307149 DOI: 10.1128/jvi.00510-20] [Citation(s) in RCA: 360] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/14/2020] [Indexed: 02/07/2023] Open
Abstract
Genetic variability across the three major histocompatibility complex (MHC) class I genes (human leukocyte antigen A [HLA-A], -B, and -C genes) may affect susceptibility to and severity of the disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus responsible for coronavirus disease 2019 (COVID-19). We performed a comprehensive in silico analysis of viral peptide-MHC class I binding affinity across 145 HLA-A, -B, and -C genotypes for all SARS-CoV-2 peptides. We further explored the potential for cross-protective immunity conferred by prior exposure to four common human coronaviruses. The SARS-CoV-2 proteome was successfully sampled and was represented by a diversity of HLA alleles. However, we found that HLA-B*46:01 had the fewest predicted binding peptides for SARS-CoV-2, suggesting that individuals with this allele may be particularly vulnerable to COVID-19, as they were previously shown to be for SARS (M. Lin, H.-T. Tseng, J. A. Trejaut, H.-L. Lee, et al., BMC Med Genet 4:9, 2003, https://bmcmedgenet.biomedcentral.com/articles/10.1186/1471-2350-4-9). Conversely, we found that HLA-B*15:03 showed the greatest capacity to present highly conserved SARS-CoV-2 peptides that are shared among common human coronaviruses, suggesting that it could enable cross-protective T-cell-based immunity. Finally, we reported global distributions of HLA types with potential epidemiological ramifications in the setting of the current pandemic.IMPORTANCE Individual genetic variation may help to explain different immune responses to a virus across a population. In particular, understanding how variation in HLA may affect the course of COVID-19 could help identify individuals at higher risk from the disease. HLA typing can be fast and inexpensive. Pairing HLA typing with COVID-19 testing where feasible could improve assessment of severity of viral disease in the population. Following the development of a vaccine against SARS-CoV-2, the virus that causes COVID-19, individuals with high-risk HLA types could be prioritized for vaccination.
Collapse
Affiliation(s)
- Austin Nguyen
- Computational Biology Program, Oregon Health & Science University, Portland, Oregon, USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Julianne K David
- Computational Biology Program, Oregon Health & Science University, Portland, Oregon, USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Sean K Maden
- Computational Biology Program, Oregon Health & Science University, Portland, Oregon, USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Mary A Wood
- Computational Biology Program, Oregon Health & Science University, Portland, Oregon, USA
- Portland VA Research Foundation, Portland, Oregon, USA
| | - Benjamin R Weeder
- Computational Biology Program, Oregon Health & Science University, Portland, Oregon, USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Abhinav Nellore
- Computational Biology Program, Oregon Health & Science University, Portland, Oregon, USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
- Department of Surgery, Oregon Health & Science University, Portland, Oregon, USA
| | - Reid F Thompson
- Computational Biology Program, Oregon Health & Science University, Portland, Oregon, USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
- Department of Radiation Medicine, Oregon Health & Science University, Portland, Oregon, USA
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon, USA
- Division of Hospital and Specialty Medicine, VA Portland Healthcare System, Portland, Oregon, USA
| |
Collapse
|
35
|
Abd Raman HS, Tan S, August JT, Khan AM. Dynamics of Influenza A (H5N1) virus protein sequence diversity. PeerJ 2020; 7:e7954. [PMID: 32518710 PMCID: PMC7261124 DOI: 10.7717/peerj.7954] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 09/26/2019] [Indexed: 11/20/2022] Open
Abstract
Background Influenza A (H5N1) virus is a global concern with potential as a pandemic threat. High sequence variability of influenza A viruses is a major challenge for effective vaccine design. A continuing goal towards this is a greater understanding of influenza A (H5N1) proteome sequence diversity in the context of the immune system (antigenic diversity), the dynamics of mutation, and effective strategies to overcome the diversity for vaccine design. Methods Herein, we report a comprehensive study of the dynamics of H5N1 mutations by analysis of the aligned overlapping nonamer positions (1–9, 2–10, etc.) of more than 13,000 protein sequences of avian and human influenza A (H5N1) viruses, reported over at least 50 years. Entropy calculations were performed on 9,408 overlapping nonamer position of the proteome to study the diversity in the context of immune system. The nonamers represent the predominant length of the binding cores for peptides recognized by the cellular immune system. To further dissect the sequence diversity, each overlapping nonamer position was quantitatively analyzed for four patterns of sequence diversity motifs: index, major, minor and unique. Results Almost all of the aligned overlapping nonamer positions of each viral proteome exhibited variants (major, minor, and unique) to the predominant index sequence. Each variant motif displayed a characteristic pattern of incidence change in relation to increased total variants. The major variant exhibited a restrictive pyramidal incidence pattern, with peak incidence at 50% total variants. Post this peak incidence, the minor variants became the predominant motif for majority of the positions. Unique variants, each sequence observed only once, were present at nearly all of the nonamer positions. The diversity motifs (index and variants) demonstrated complex inter-relationships, with motif switching being a common phenomenon. Additionally, 25 highly conserved sequences were identified to be shared across viruses of both hosts, with half conserved to several other influenza A subtypes. Discussion The presence of distinct sequences (nonatypes) at nearly all nonamer positions represents a large repertoire of reported viral variants in the proteome, which influence the variability dynamics of the viral population. This work elucidated and provided important insights on the components that make up the viral diversity, delineating inherent patterns in the organization of sequence changes that function in the viral fitness-selection. Additionally, it provides a catalogue of all the mutational changes involved in the dynamics of H5N1 viral diversity for both avian and human host populations. This work provides data relevant for the design of prophylactics and therapeutics that overcome the diversity of the virus, and can aid in the surveillance of existing and future strains of influenza viruses.
Collapse
Affiliation(s)
| | - Swan Tan
- School of Data Sciences, Perdana University, Serdang, Selangor, Malaysia.,Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, United States of America
| | - Joseph Thomas August
- School of Medicine, Johns Hopkins University, Baltimore, MD, United States of America
| | - Asif M Khan
- School of Data Sciences, Perdana University, Serdang, Selangor, Malaysia.,School of Medicine, Johns Hopkins University, Baltimore, MD, United States of America.,Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Beykoz, Istanbul, Turkey
| |
Collapse
|
36
|
Leng Q, Tarbe M, Long Q, Wang F. Pre-existing heterologous T-cell immunity and neoantigen immunogenicity. Clin Transl Immunology 2020; 9:e01111. [PMID: 32211191 PMCID: PMC7085466 DOI: 10.1002/cti2.1111] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/13/2020] [Accepted: 01/30/2020] [Indexed: 12/22/2022] Open
Abstract
Neoantigens are tumor‐specific mutated proteins that are exempt from central tolerance and are therefore capable of efficiently eliciting effective T‐cell responses. The identification of immunogenic neoantigens in tumor‐specific mutated proteins has promising clinical implications for cancer immunotherapy. However, the factors that may contribute to neoantigen immunogenicity are not yet fully understood. Through molecular mimicry of antigens arising during cancer progression, the gut microbiota and previously encountered pathogens potentially have profound impacts on T‐cell responses to previously unencountered tumor neoantigens. Here, we review the characteristics of immunogenic neoantigens and how host exposure to microbes may affect T‐cell responses to neoantigens. We address the hypothesis that pre‐existing heterologous memory T‐cell immunity is a major factor that influences neoantigen immunogenicity in individual cancer patients. Accumulating data suggest that differences in individual histories of microbial exposure should be taken into account during the optimisation of algorithms that predict neoantigen immunogenicity.
Collapse
Affiliation(s)
- Qibin Leng
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University State Key Laboratory of Respiratory Diseases, Guangzhou Medical University Guangzhou China.,The Joint Center for Infection and Immunity Guangzhou Women and Children's Medical Center Guangzhou Institute of Pediatrics Guangzhou Medical University Guangzhou China.,Institute Pasteur of Shanghai Chinese Academy of Science Shanghai China
| | - Marion Tarbe
- The Joint Center for Infection and Immunity Guangzhou Women and Children's Medical Center Guangzhou Institute of Pediatrics Guangzhou Medical University Guangzhou China.,Institute Pasteur of Shanghai Chinese Academy of Science Shanghai China
| | - Qi Long
- Department of Biostatistics, Epidemiology and Informatics Perelman School of Medicine University of Pennsylvania Philadelphia PA USA
| | - Feng Wang
- Department of Immunology and Microbiology Center for Microbiota and Immunological Diseases Shanghai General Hospital Shanghai Institute of Immunology Shanghai Jiao Tong University School of Medicine Shanghai China.,Research Center of Translational Medicine Shanghai Children's Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| |
Collapse
|
37
|
Meilleur CE, Wardell CM, Mele TS, Dikeakos JD, Bennink JR, Mu HH, McCormick JK, Haeryfar SMM. Bacterial Superantigens Expand and Activate, Rather than Delete or Incapacitate, Preexisting Antigen-Specific Memory CD8+ T Cells. J Infect Dis 2020; 219:1307-1317. [PMID: 30418594 DOI: 10.1093/infdis/jiy647] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/07/2018] [Indexed: 11/13/2022] Open
Abstract
Superantigens (SAgs) released by common Gram-positive bacterial pathogens have been reported to delete, anergize, or activate mouse T cells. However, little is known about their effects on preexisting memory CD8+ T cell (TCD8) pools. Furthermore, whether SAgs manipulate human memory TCD8 responses to cognate antigens is unknown. We used a human peripheral blood mononuclear cell culture system and a nontransgenic mouse model in which the impact of stimulation by two fundamentally distinct SAgs, staphylococcal enterotoxin B and Mycoplasma arthritidis mitogen, on influenza virus- and/or cytomegalovirus-specific memory TCD8 could be monitored. Bacterial SAgs surprisingly expanded antiviral memory TCD8 generated naturally through infection or artificially through vaccination. Mechanistically, this was a T cell-intrinsic and T cell receptor β-chain variable-dependent phenomenon. Importantly, SAg-expanded TCD8 displayed an effector memory phenotype and were capable of producing interferon-γ and destroying target cells ex vivo or in vivo. These findings have clear implications for antimicrobial defense and rational vaccine design.
Collapse
Affiliation(s)
- Courtney E Meilleur
- Department of Microbiology and Immunology, Western University, London, Canada
| | - Christine M Wardell
- Department of Microbiology and Immunology, Western University, London, Canada
| | - Tina S Mele
- Division of General Surgery, Department of Surgery, Western University, London, Canada.,Division of Critical Care Medicine, Western University, London, Canada
| | - Jimmy D Dikeakos
- Department of Microbiology and Immunology, Western University, London, Canada
| | - Jack R Bennink
- Viral Immunology Section, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Hong-Hua Mu
- Division of Rheumatology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City
| | - John K McCormick
- Department of Microbiology and Immunology, Western University, London, Canada.,Centre for Human Immunology, Western University, London, Canada.,Lawson Health Research Institute, London, Canada
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, London, Canada.,Division of General Surgery, Department of Surgery, Western University, London, Canada.,Division of Clinical Immunology and Allergy, Department of Medicine, Western University, London, Canada.,Centre for Human Immunology, Western University, London, Canada.,Lawson Health Research Institute, London, Canada
| |
Collapse
|
38
|
Bolek KJ, Klasing KC. The effects of vaccination with keyhole limpet hemocyanin or oral administration of Salmonella enterica serovar Enteritidis on the growth performance of immunoglobulin knockout chickens. Poult Sci 2019; 98:3504-3513. [PMID: 31329978 DOI: 10.3382/ps/pez172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 03/21/2019] [Indexed: 11/20/2022] Open
Abstract
The nutritional cost of activating B cell-mediated immunity is thought to be low in chickens. However, this assumption is incompletely characterized. Immunoglobulin knockout (Ig-KO) chickens lacking B cells and immunoglobulin may potentially be a robust model to investigate the nutritional cost of immunity. Specifically, comparing the growth of Ig-KO chickens to immunocompetent birds (WT) following activation of the immune system should indicate costs associated with B cell-mediated immunity. A total of 3 experiments were conducted to determine if (1) an antibody response to keyhole limpet hemocyanin (KLH), (2) an intra-abdominal injection with Salmonella enterica serovar Enteritidis (SE), or (3) an oral challenge of SE would affect body weight gain (BWG) in homozygous Ig-KO (KO) chickens and their immunocompetent counterparts. In Experiment 1, a significant genotype*vaccination status interaction was observed (P = 0.03) during the period from 0 to 6 D after initial vaccination in which KLH injection reduced BWG in WT birds, but not in KO birds. A genotype*vaccination status interaction was present (P = 0.002) from 0 to 7 D after the first booster in which KLH injection due to reduced BWG in KO birds, but not WT birds. In Experiment 2, both KO and immunocompetent birds lost body weight during the period from 0 to 2 D after the SE injection, with no significant differences due to genotype (P = 0.92). Experiment 3 demonstrated that KO birds gained less weight than immunocompetent birds, with a tendency for less weight gain after an initial challenge (P = 0.07) and significantly so after a secondary challenge (P = 0.03). The results from these experiments collectively demonstrate that B cell-mediated immunity can affect growth performance in chickens. Furthermore, these effects can either preserve or impair growth performance, likely via mechanisms related to the immune response rather than the discrete nutritional cost of B cell-mediated immunity.
Collapse
Affiliation(s)
- K J Bolek
- Department of Animal Science, University of California, Davis, CA 95616
| | - K C Klasing
- Department of Animal Science, University of California, Davis, CA 95616
| |
Collapse
|
39
|
Pardy RD, Richer MJ. Protective to a T: The Role of T Cells during Zika Virus Infection. Cells 2019; 8:cells8080820. [PMID: 31382545 PMCID: PMC6721718 DOI: 10.3390/cells8080820] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 07/31/2019] [Accepted: 08/02/2019] [Indexed: 12/22/2022] Open
Abstract
CD4 and CD8 T cells are an important part of the host's capacity to defend itself against viral infections. During flavivirus infections, T cells have been implicated in both protective and pathogenic responses. Given the recent emergence of Zika virus (ZIKV) as a prominent global health threat, the question remains as to how T cells contribute to anti-ZIKV immunity. Furthermore, high homology between ZIKV and other, co-circulating flaviviruses opens the possibility of positive or negative effects of cross-reactivity due to pre-existing immunity. In this review, we will discuss the CD4 and CD8 T cell responses to ZIKV, and the lessons we have learned from both mouse and human infections. In addition, we will consider the possibility of whether T cells, in the context of flavivirus-naïve and flavivirus-immune subjects, play a role in promoting ZIKV pathogenesis during infection.
Collapse
Affiliation(s)
- Ryan D Pardy
- Department of Microbiology & Immunology, McGill University, Montreal, QC H3A 2B4, Canada
- Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Martin J Richer
- Department of Microbiology & Immunology, McGill University, Montreal, QC H3A 2B4, Canada.
- Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal, QC H3G 1Y6, Canada.
| |
Collapse
|
40
|
Quantification of epitope abundance reveals the effect of direct and cross-presentation on influenza CTL responses. Nat Commun 2019; 10:2846. [PMID: 31253788 PMCID: PMC6599079 DOI: 10.1038/s41467-019-10661-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 05/24/2019] [Indexed: 11/08/2022] Open
Abstract
The magnitude of T cell responses to infection is a function of the naïve T cell repertoire combined with the context and duration of antigen presentation. Using mass spectrometry, we identify and quantify 21 class 1 MHC-restricted influenza A virus (IAV)-peptides following either direct or cross-presentation. All these peptides, including seven novel epitopes, elicit T cell responses in infected C57BL/6 mice. Directly presented IAV epitopes maintain their relative abundance across distinct cell types and reveal a broad range of epitope abundances. In contrast, cross-presented epitopes are more uniform in abundance. We observe a clear disparity in the abundance of the two key immunodominant IAV antigens, wherein direct infection drives optimal nucleoprotein (NP)366–374 presentation, while cross-presentation is optimal for acid polymerase (PA)224–233 presentation. The study demonstrates how assessment of epitope abundance in both modes of antigen presentation is necessary to fully understand the immunogenicity and response magnitude to T cell epitopes. CTL responses are critical in protection against pathogens. Here, using mass spectrometry and flow cytometry, the authors characterize the kinetics of influenza A virus class I MHC epitopes cross-presented in professional antigen presenting cells and identify new epitopes that elicit T cell responses in infected mice.
Collapse
|
41
|
Cipitelli MDC, Amâncio Paiva I, Badolato-Corrêa J, de-Oliveira-Pinto LM. Influence of chemokines on the endothelial permeability and cellular transmigration during dengue. Immunol Lett 2019; 212:88-97. [PMID: 31181280 DOI: 10.1016/j.imlet.2019.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/24/2019] [Accepted: 06/06/2019] [Indexed: 01/31/2023]
Abstract
During a pathogenic infection, an inflammatory process is triggered in which several inflammatory mediators, such as cytokines, chemokines, growth factors, complement system components, nitric oxide, and others induce integrity alteration on the endothelial barrier. Chemokines are responsible for regulating leukocyte trafficking under homeostatic conditions as well as activating immune system cells under inflammatory conditions. They are crucial molecules in the early stages of infection, leading to the recruitment of immune cells, namely neutrophils, monocytes, natural killer (NK) cells, natural killer T cells (NKT), dendritic cells (DC), T lymphocytes and all cells expressing chemokine receptors for inflammatory sites. Other functions, such as collagen production, tissue repair, a proliferation of hematopoietic precursors and angiogenesis, are also performed by these molecules. Chemokines, amongst inflammatory mediators, play a key role in dengue immunopathogenesis. Dengue fever is a disease caused by the dengue virus (DENV). It is characterized by a broad spectrum of clinical manifestations ranging from asymptomatic cases to mild and severe symptomatic ones. As for the latter, the appearance of hemorrhagic manifestations and changes in vascular permeability may lead the patient to develop cavitary effusions, organ involvement, and even death. As chemokines exert an influence on various homeostatic and inflammatory processes, acting vigorously on vascular endothelial activation and cell migration, the main purpose of this chapter is to discuss the influence of chemokines on the alteration of endothelial permeability and migration of T lymphocytes in DENV infection.
Collapse
Affiliation(s)
- Márcio da Costa Cipitelli
- Laboratory of Viral Immunology, Oswaldo Cruz Institute, Oswaldo Cruz Fundation, Rio de Janeiro, Brazil
| | - Iury Amâncio Paiva
- Laboratory of Viral Immunology, Oswaldo Cruz Institute, Oswaldo Cruz Fundation, Rio de Janeiro, Brazil
| | - Jéssica Badolato-Corrêa
- Laboratory of Viral Immunology, Oswaldo Cruz Institute, Oswaldo Cruz Fundation, Rio de Janeiro, Brazil
| | | |
Collapse
|
42
|
Nienen M, Stervbo U, Mölder F, Kaliszczyk S, Kuchenbecker L, Gayova L, Schweiger B, Jürchott K, Hecht J, Neumann AU, Rahmann S, Westhoff T, Reinke P, Thiel A, Babel N. The Role of Pre-existing Cross-Reactive Central Memory CD4 T-Cells in Vaccination With Previously Unseen Influenza Strains. Front Immunol 2019; 10:593. [PMID: 31019503 PMCID: PMC6458262 DOI: 10.3389/fimmu.2019.00593] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/05/2019] [Indexed: 11/13/2022] Open
Abstract
Influenza vaccination is a common approach to prevent seasonal and pandemic influenza. Pre-existing antibodies against close viral strains might impair antibody formation against previously unseen strains-a process called original antigenic sin. The role of this pre-existing cellular immunity in this process is, despite some hints from animal models, not clear. Here, we analyzed cellular and humoral immunity in healthy individuals before and after vaccination with seasonal influenza vaccine. Based on influenza-specific hemagglutination inhibiting (HI) titers, vaccinees were grouped into HI-negative and -positive cohorts followed by in-depth cytometric and TCR repertoire analysis. Both serological groups revealed cross-reactive T-cell memory to the vaccine strains at baseline that gave rise to the majority of vaccine-specific T-cells post vaccination. On the contrary, very limited number of vaccine-specific T-cell clones was recruited from the naive pool. Furthermore, baseline quantity of vaccine-specific central memory helper T-cells and clonotype richness of this population directly correlated with the vaccination efficacy. Our findings suggest that the deliberate recruitment of pre-existing cross-reactive cellular memory might help to improve vaccination outcome.
Collapse
Affiliation(s)
- Mikalai Nienen
- Institute for Medical Immunology, Charité University Medicine Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine Berlin, Berlin, Germany.,Labor Berlin-Charité Vivantes GmbH, Berlin, Germany
| | - Ulrik Stervbo
- Center for Translational Medicine, Immunology and Transplantation, Marien Hospital Herne, Ruhr University Bochum, Herne, Germany
| | - Felix Mölder
- Genome Informatics, Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sviatlana Kaliszczyk
- Center for Translational Medicine, Immunology and Transplantation, Marien Hospital Herne, Ruhr University Bochum, Herne, Germany
| | | | | | | | - Karsten Jürchott
- Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine Berlin, Berlin, Germany
| | - Jochen Hecht
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Avidan U Neumann
- Institute of Environmental Medicine, German Research Center for Environmental Health, Helmholtz Zentrum München, Augsburg, Germany
| | - Sven Rahmann
- Genome Informatics, Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Timm Westhoff
- Department of Internal Medicine, Marien Hospital Herne, Ruhr University Bochum, Herne, Germany
| | - Petra Reinke
- Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine Berlin, Berlin, Germany.,Department of Nephrology and Intensive Care, Charité University Medicine Berlin, Berlin, Germany
| | - Andreas Thiel
- Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine Berlin, Berlin, Germany
| | - Nina Babel
- Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine Berlin, Berlin, Germany.,Center for Translational Medicine, Immunology and Transplantation, Marien Hospital Herne, Ruhr University Bochum, Herne, Germany.,Department of Nephrology and Intensive Care, Charité University Medicine Berlin, Berlin, Germany
| |
Collapse
|
43
|
Heide J, Vaughan KC, Sette A, Jacobs T, Schulze Zur Wiesch J. Comprehensive Review of Human Plasmodium falciparum-Specific CD8+ T Cell Epitopes. Front Immunol 2019; 10:397. [PMID: 30949162 PMCID: PMC6438266 DOI: 10.3389/fimmu.2019.00397] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/15/2019] [Indexed: 12/16/2022] Open
Abstract
Control of malaria is an important global health issue and there is still an urgent need for the development of an effective prophylactic vaccine. Multiple studies have provided strong evidence that Plasmodium falciparum-specific MHC class I-restricted CD8+ T cells are important for sterile protection against Plasmodium falciparum infection. Here, we present an interactive epitope map of all P. falciparum-specific CD8+ T cell epitopes published to date, based on a comprehensive data base (IEDB), and literature search. The majority of the described P. falciparum-specific CD8+ T cells were directed against the antigens CSP, TRAP, AMA1, and LSA1. Notably, most of the epitopes were discovered in vaccine trials conducted with malaria-naïve volunteers. Only few immunological studies of P. falciparum-specific CD8+ T cell epitopes detected in patients suffering from acute malaria or in people living in malaria endemic areas have been published. Further detailed immunological mappings of P. falciparum-specific epitopes of a broader range of P. falciparum proteins in different settings and with different disease status are needed to gain a more comprehensive understanding of the role of CD8+ T cell responses for protection, and to better guide vaccine design and to study their efficacy.
Collapse
Affiliation(s)
- Janna Heide
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Kerrie C Vaughan
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States.,Department of Medicine, Division of Infectious Diseases, University of California, San Diego (UCSD), La Jolla, CA, United States
| | - Thomas Jacobs
- Protozoa Immunology, Bernhard-Nocht-Institute of Tropical Medicine, Hamburg, Germany
| | - Julian Schulze Zur Wiesch
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| |
Collapse
|
44
|
Kolawole EM, Andargachew R, Liu B, Jacobs JR, Evavold BD. 2D Kinetic Analysis of TCR and CD8 Coreceptor for LCMV GP33 Epitopes. Front Immunol 2018; 9:2348. [PMID: 30374353 PMCID: PMC6197077 DOI: 10.3389/fimmu.2018.02348] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/21/2018] [Indexed: 11/13/2022] Open
Abstract
The LCMV GP33 CD8 epitope has long been one of the most widely used antigens in viral immunology. Of note, almost all of the in vitro analyses of CD8 T cell responses to this epitope make use of an altered peptide ligand (APL) in which the cysteine from the original 9-mer peptide (KAVYNFATC) is substituted by a methionine at position 41 (KAVYNFATM). In addition, it is possible that the antigen processed during natural LCMV infection is an 11-mer peptide (KAVYNFATCGI) rather than the widely used 9-mer. Although previous affinity measurements using purified proteins for these antigen variants revealed minimal differences, we applied highly sensitive two dimensional (2D) biophysical based techniques to further dissect TCR interaction with these closely related GP33 variants. The kinetic analyses of affinity provided by the 2D micropipette adhesion frequency assay (2D-MP) and bond lifetime under force analyzed using a biomembrane force probe (BFP) revealed significant differences between 41M, 41C and the 11-mer 41CGI antigen. We found a hierarchy in 2D affinity as 41M peptide displayed augmented TCR 2D affinity compared to 41C and 41CGI. These differences were also maintained in the presence of CD8 coreceptor and when analysis of total TCR:pMHC and CD8:pMHC bonds were considered. Moreover, the three ligands displayed dramatic differences in the bond lifetimes generated under force, in particular the 41CGI variant with the lowest 2D affinity demonstrated a 15-fold synergistic contribution of the CD8 coreceptor to overall bond lifetime. Our analyses emphasize the sensitivity of single cell and single bond 2D kinetic measurements in distinguishing between related agonist peptides.
Collapse
Affiliation(s)
- Elizabeth M Kolawole
- Department of Pathology, Microbiology and Immunology, University of Utah, Salt Lake City, UT, United States
| | - Rakieb Andargachew
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, United States
| | - Baoyu Liu
- Department of Pathology, Microbiology and Immunology, University of Utah, Salt Lake City, UT, United States
| | - Jesica R Jacobs
- Department of Pathology, Microbiology and Immunology, University of Utah, Salt Lake City, UT, United States
| | - Brian D Evavold
- Department of Pathology, Microbiology and Immunology, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
45
|
Lim MQ, Kumaran EAP, Tan HC, Lye DC, Leo YS, Ooi EE, MacAry PA, Bertoletti A, Rivino L. Cross-Reactivity and Anti-viral Function of Dengue Capsid and NS3-Specific Memory T Cells Toward Zika Virus. Front Immunol 2018; 9:2225. [PMID: 30327651 PMCID: PMC6174860 DOI: 10.3389/fimmu.2018.02225] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 09/07/2018] [Indexed: 12/01/2022] Open
Abstract
Zika virus (ZIKV), a flavivirus with homology to dengue virus (DENV), is spreading to areas of DENV hyper-endemicity. Heterologous T cell immunity, whereby virus-specific memory T cells are activated by variant peptides derived from a different virus, can lead to enhanced viral clearance or diminished protective immunity and altered immunopathology. In mice, CD8+ T cells specific for DENV provide in vivo protective efficacy against subsequent ZIKV infection. In humans, contrasting studies report complete absence or varying degrees of DENV/ZIKV T cell cross-reactivity. Moreover, the impact of cross-reactive T cell recognition on the anti-viral capacity of T cells remains unclear. Here, we show that DENV-specific memory T cells display robust cross-reactive recognition of ZIKV NS3 ex vivo and after in vitro expansion in respectively n = 7/10 and n = 9/9 dengue-immune individuals tested. In contrast, cross-reactivity toward ZIKV capsid is low or absent. Cross-reactive recognition of DENV or ZIKV NS3 peptides elicits similar production of the anti-viral effector mediators IFN-γ, TNF-α, and CD107a. We identify 9 DENV/ZIKV cross-reactive epitopes, 7 of which are CD4+ and 2 are CD8+ T cell epitopes. We also show that cross-reactive CD4+ and CD8+ T cells targeting novel NS3 epitopes display anti-viral effector potential toward ZIKV-infected cells, with CD8+ T cells mediating direct lyses of these cells. Our results demonstrate that DENV NS3-specific memory T cells display anti-viral effector capacity toward ZIKV, suggesting a potential beneficial effect in humans of pre-existing T cell immunity to DENV upon ZIKV infection.
Collapse
Affiliation(s)
- Mei Qiu Lim
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Emmanuelle A P Kumaran
- Immunology Programme, Department of Microbiology and Immunology, Life Science Institute, National University of Singapore, Singapore, Singapore
| | - Hwee Cheng Tan
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - David C Lye
- Communicable Disease Centre, Institute of Infectious Disease and Epidemiology, Tan Tock Seng Hospital, Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yee Sin Leo
- Communicable Disease Centre, Institute of Infectious Disease and Epidemiology, Tan Tock Seng Hospital, Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,National Centre for Infectious diseases, NCID, Singapore, Singapore
| | - Eng Eong Ooi
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore.,Immunology Programme, Department of Microbiology and Immunology, Life Science Institute, National University of Singapore, Singapore, Singapore
| | - Paul A MacAry
- Immunology Programme, Department of Microbiology and Immunology, Life Science Institute, National University of Singapore, Singapore, Singapore
| | - Antonio Bertoletti
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore.,Singapore Immunology Network, Singapore Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Laura Rivino
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
46
|
Kim J, De La Cruz J, Lam K, Ng H, Daar ES, Balamurugan A, Yang OO. CD8 + Cytotoxic T Lymphocyte Responses and Viral Epitope Escape in Acute HIV-1 Infection. Viral Immunol 2018; 31:525-536. [PMID: 30059271 DOI: 10.1089/vim.2018.0040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Epitope escape from HIV-1-targeted CD8+ cytotoxic T lymphocyte (CTL) responses occurs rapidly after acute infection and contributes to the eventual failure of effective immune control of HIV-1 infection. Because the early CTL response is key in determining HIV-1 disease outcome, studying the process of epitope escape is crucial for understanding what leads to failure of immune control in acute HIV-1 infection and will provide important implications for HIV-1 vaccine design. HIV-1-specific CD8+ T lymphocyte responses against viral epitopes were mapped in six acutely infected individuals, and the magnitudes of these responses were measured longitudinally during acute infection. The evolution of autologous circulating viral epitopes was determined in four of these subjects. In-depth testing of CD8+ T lymphocyte responses against index and all autologous-detected variant epitopes was performed in one subject. Among the four individuals examined, 10 of a total of 35 CD8+ T cell responses within Gag, Pol, and Nef showed evidence of epitope escape. CTL responses with viral epitope variant evolution were shown in one subject, and this evolution occurred with and without measurable CTL responses against epitope variants. These results demonstrate a dynamic period of viral epitope evolution in early HIV-1 infection due to CD8+ CTL response pressure.
Collapse
Affiliation(s)
- Joseph Kim
- 1 Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California , Los Angeles, California
| | - Justin De La Cruz
- 1 Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California , Los Angeles, California
| | - Karen Lam
- 2 Department of Microbiology, Immunology, and Molecular Genetics, University of California , Los Angeles, California
| | - Hwee Ng
- 1 Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California , Los Angeles, California
| | - Eric S Daar
- 3 Los Angeles Biomedical Research Institute , Harbor-UCLA Medical Center, Torrance, California
| | - Arumugam Balamurugan
- 1 Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California , Los Angeles, California
| | - Otto O Yang
- 1 Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California , Los Angeles, California.,2 Department of Microbiology, Immunology, and Molecular Genetics, University of California , Los Angeles, California.,4 AIDS Healthcare Foundation , Los Angeles, California
| |
Collapse
|
47
|
Understanding the Human T Cell Response to Dengue Virus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1062:241-250. [PMID: 29845537 DOI: 10.1007/978-981-10-8727-1_17] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Our understanding of how T cells respond to dengue virus has greatly advanced in the last decade but important questions still remain unanswered. Dengue virus infection elicits a broad anti-viral T cell response with NS3, NS4b and NS5 being the main targets for CD8+ T cells, which dominate the response while the structural proteins capsid, envelope and the secreted protein NS1 are the preferential targets for CD4+ T cells. Upon T cell activation during acute dengue infection, dengue-specific T cells acquire expression of the skin-homing marker cutaneous associated antigen (CLA) and they can be found at high frequencies in the skin of infected patients. This suggests that the skin represents an important site for the immuno surveillance of dengue virus. The immunoprotective role of skin-homing dengue-specific T cells, their potential involvement in pathological skin manifestations and their long-term persistence as tissue resident T cells to provide immediate onsite protection are open questions that we are currently investigating. The contribution of pre-existing dengue-specific T cells towards protective immunity and/or immunopathology during secondary dengue infection remains a major knowledge gap. The evidence supporting these opposing outcomes and our current understanding of the characteristics of the human T cell response to dengue virus will be discussed.
Collapse
|
48
|
Zhao M, Zhang H, Liu K, Gao GF, Liu WJ. Human T-cell immunity against the emerging and re-emerging viruses. SCIENCE CHINA. LIFE SCIENCES 2017; 60:1307-1316. [PMID: 29294219 PMCID: PMC7089170 DOI: 10.1007/s11427-017-9241-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 11/12/2017] [Indexed: 12/21/2022]
Abstract
Over the past decade, we have seen an alarming number of high-profile outbreaks of newly emerging and re-emerging viruses. Recent outbreaks of avian influenza viruses, Middle East respiratory syndrome coronaviruses, Zika virus and Ebola virus present great threats to global health. Considering the pivotal role of host T-cell immunity in the alleviation of symptoms and the clearance of viruses in patients, there are three issues to be primarily concerned about T-cell immunity when a new virus emerges: first, does the population possess pre-existing T-cells against the new virus through previous infections of genetically relevant viruses; second, does a proper immune response arise in the patients to provide protection through an immunopathogenic effect; lastly, how long can the virus-specific immune memory persist. Herein, we summarize the current updates on the characteristics of human T-cell immunological responses against recently emerged or re-emerged viruses, and emphasize the necessity for timely investigation on the T-cell features of these viral diseases, which may provide beneficial recommendations for clinical diagnosis and vaccine development.
Collapse
Affiliation(s)
- Min Zhao
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, 100101, China
| | - Hangjie Zhang
- Key Laboratory of Medical Virology and Viral Diseases, Ministry of Health of People's Republic of China, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Kefang Liu
- Key Laboratory of Medical Virology and Viral Diseases, Ministry of Health of People's Republic of China, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - George F Gao
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, 100101, China
- Key Laboratory of Medical Virology and Viral Diseases, Ministry of Health of People's Republic of China, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - William J Liu
- Key Laboratory of Medical Virology and Viral Diseases, Ministry of Health of People's Republic of China, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China.
| |
Collapse
|
49
|
Epitope-Specific Vaccination Limits Clonal Expansion of Heterologous Naive T Cells during Viral Challenge. Cell Rep 2017; 17:636-644. [PMID: 27732841 DOI: 10.1016/j.celrep.2016.09.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 06/21/2016] [Accepted: 09/06/2016] [Indexed: 11/20/2022] Open
Abstract
Despite robust secondary T cell expansion primed by vaccination, the impact on primary immune responses to heterotypic antigens remains undefined. Here we show that secondary expansion of epitope-specific memory CD8+ T cells primed by prior infection with recombinant pathogens limits the primary expansion of naive CD8+ T cells with specificity to new heterologous antigens, dampening protective immunity against subsequent pathogen challenge. The degree of naive T cell repression directly paralleled the magnitude of the recall response. Suppressed primary T cell priming reflects competition for antigen accessibility, since clonal expansion was not inhibited if the primary and secondary epitopes were expressed on different dendritic cells. Interestingly, robust recall responses did not impact antigen-specific NK cells, suggesting that adaptive and innate lymphocyte responses possess different activation requirements or occur in distinct anatomical locations. These findings have important implications in pathogen vaccination strategies that depend on the targeting of multiple T cell epitopes.
Collapse
|
50
|
Blevins S, Huseby ES. Killer T cells with a beta-flavi(r) for dengue. Nat Immunol 2017; 18:1186-1188. [PMID: 29044243 DOI: 10.1038/ni.3833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Sydney Blevins
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Eric S Huseby
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|