1
|
Ai J, Xiao H, Zhang L, Ma H, Wang D, Dilmurat D, Wang R, Xie Z. Dysfunction of γδ T cells in pediatric chronic active Epstein-Barr virus infection. Ital J Pediatr 2024; 50:213. [PMID: 39396033 PMCID: PMC11470581 DOI: 10.1186/s13052-024-01783-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/02/2024] [Indexed: 10/14/2024] Open
Abstract
Chronic active Epstein-Barr virus infection (CAEBV) is a progressive and life-threatening disease characterized by persistent or recurrent EBV activation. It has been reported that, γδ T cells, a type of cytotoxic lymphocyte, play a critical role in restricting EBV. However, the functional status of γδ T cells in pediatric CAEBV patients has not yet been described. In this study, flow cytometry analysis was conducted to explore the cytokine production capacity of γδ T cells in CAEBV patients. A diminished frequency of γδ T cells and decreased expression of cytolytic molecule granzyme B were found in CAEBV patients, suggesting a dysfunction in the immune regulatory function of γδ T cells in this disease.
Collapse
Affiliation(s)
- Junhong Ai
- Laboratory of Infection and Virology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
- Research Unit of Critical Infection in Children, 2019RU016, Chinese Academy of Medical Sciences, Beijing, 100045, China
| | - Haijuan Xiao
- Department of Infectious Diseases, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Linlin Zhang
- Laboratory of Infection and Virology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
- Research Unit of Critical Infection in Children, 2019RU016, Chinese Academy of Medical Sciences, Beijing, 100045, China
- Medical Oncology Department, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Honghao Ma
- Hematology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Dong Wang
- Hematology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Dilara Dilmurat
- Laboratory of Infection and Virology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
- Research Unit of Critical Infection in Children, 2019RU016, Chinese Academy of Medical Sciences, Beijing, 100045, China
| | - Ran Wang
- Laboratory of Infection and Virology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
- Research Unit of Critical Infection in Children, 2019RU016, Chinese Academy of Medical Sciences, Beijing, 100045, China.
| | - Zhengde Xie
- Laboratory of Infection and Virology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
- Research Unit of Critical Infection in Children, 2019RU016, Chinese Academy of Medical Sciences, Beijing, 100045, China.
| |
Collapse
|
2
|
Yang Y, Ding T, Cong Y, Luo X, Liu C, Gong T, Zhao M, Zheng X, Li C, Zhang Y, Zhou J, Ni C, Zhang X, Ji Z, Wu T, Yang S, Zhou Q, Wu D, Gong X, Zheng Q, Li X. Interferon-induced transmembrane protein-1 competitively blocks Ephrin receptor A2-mediated Epstein-Barr virus entry into epithelial cells. Nat Microbiol 2024; 9:1256-1270. [PMID: 38649412 PMCID: PMC11087256 DOI: 10.1038/s41564-024-01659-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 03/04/2024] [Indexed: 04/25/2024]
Abstract
Epstein-Barr virus (EBV) can infect both B cells and epithelial cells (ECs), causing diseases such as mononucleosis and cancer. It enters ECs via Ephrin receptor A2 (EphA2). The function of interferon-induced transmembrane protein-1 (IFITM1) in EBV infection of ECs remains elusive. Here we report that IFITM1 inhibits EphA2-mediated EBV entry into ECs. RNA-sequencing and clinical sample analysis show reduced IFITM1 in EBV-positive ECs and a negative correlation between IFITM1 level and EBV copy number. IFITM1 depletion increases EBV infection and vice versa. Exogenous soluble IFITM1 effectively prevents EBV infection in vitro and in vivo. Furthermore, three-dimensional structure prediction and site-directed mutagenesis demonstrate that IFITM1 interacts with EphA2 via its two specific residues, competitively blocking EphA2 binding to EBV glycoproteins. Finally, YTHDF3, an m6A reader, suppresses IFITM1 via degradation-related DEAD-box protein 5 (DDX5). Thus, this study underscores IFITM1's crucial role in blocking EphA2-mediated EBV entry into ECs, indicating its potential in preventing EBV infection.
Collapse
Affiliation(s)
- Yinggui Yang
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Tengteng Ding
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Ying Cong
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaomin Luo
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Changlin Liu
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Ting Gong
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Min Zhao
- PANACRO(Hefei) Pharmaceutical Technology Co. Ltd., Hefei, China
| | - Xichun Zheng
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Chenglin Li
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Yuanbin Zhang
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiayi Zhou
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Chuping Ni
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Xueyu Zhang
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Ziliang Ji
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Tao Wu
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Shaodong Yang
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Qingchun Zhou
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Dinglan Wu
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China.
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| | - Xinqi Gong
- Mathematical Intelligence Application LAB, Institute for Mathematical Sciences, Renmin University of China, Beijing, China.
| | - Qingyou Zheng
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China.
| | - Xin Li
- Shenzhen Key Laboratory of Viral Oncology, Department of Urology, and Clinical Innovation and Research Centre (CIRC), Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China.
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
3
|
Gutiérrez-Guerrero A, Espinosa-Padilla SE, Lugo-Reyes SO. [Anything that can go wrong: cytotoxic cells and their control of Epstein-Barr virus]. REVISTA ALERGIA MÉXICO 2024; 71:29-39. [PMID: 38683066 DOI: 10.29262/ram.v71i1.1276] [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: 06/30/2023] [Accepted: 08/31/2023] [Indexed: 05/01/2024] Open
Abstract
Epstein-Barr virus (EBV) is an gamma of herpes virus affecting exclusively humans, was the first oncogenic virus described and is associated with over seven different cancers. Curiously, the exchange of genes during viral infections has enabled the evolution of other cellular organisms, favoring new functions and the survival of the host. EBV has been co-evolving with mammals for hundreds of millions of years, and more than 95% of adults have been infected in one moment of their life. The infection is acquired primarily during childhood, in most cases as an asymptomatic infection. However, during adolescence or young adulthood, around 10 to 30% develop infectious mononucleosis. The NK and CD8+ T cells are the cytotoxic cells of the immune system that focus on antiviral responses. Importantly, an essential role of NK and CD8+ T cells has been demonstrated during the control and elimination of EBV-infected cells. Nonetheless, when the cytotoxic function of these cells is compromised, the infection increases the risk of developing lymphoproliferative diseases and cancer, often fatal. In this review, we delineate EBV infection and the importance of cytotoxic responses by NK and CD8+ T cells during the control and elimination of EBV-infected cells. Furthermore, we briefly discuss the main inborn errors of immunity that compromise cytotoxic responses by NK and CD8+ T cells, and how this scenario affects the antiviral response during EBV infection. Finally, we conclude the review by underlying the need for an effective EBV vaccine capable of preventing infection and the consequent development of malignancies and autoimmune diseases.
Collapse
Affiliation(s)
- Arturo Gutiérrez-Guerrero
- Laboratorio de Inmunodeficiencias, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México
| | - Sara Elva Espinosa-Padilla
- Laboratorio de Inmunodeficiencias, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México
| | - Saúl Oswaldo Lugo-Reyes
- Laboratorio de Inmunodeficiencias, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México
| |
Collapse
|
4
|
Zhang Q, Xu M. EBV-induced T-cell responses in EBV-specific and nonspecific cancers. Front Immunol 2023; 14:1250946. [PMID: 37841280 PMCID: PMC10576448 DOI: 10.3389/fimmu.2023.1250946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/12/2023] [Indexed: 10/17/2023] Open
Abstract
Epstein-Barr virus (EBV) is a ubiquitous human tumor virus associated with various malignancies, including B-lymphoma, NK and T-lymphoma, and epithelial carcinoma. It infects B lymphocytes and epithelial cells within the oropharynx and establishes persistent infection in memory B cells. With a balanced virus-host interaction, most individuals carry EBV asymptomatically because of the lifelong surveillance by T cell immunity against EBV. A stable anti-EBV T cell repertoire is maintained in memory at high frequency in the blood throughout persistent EBV infection. Patients with impaired T cell immunity are more likely to develop life-threatening lymphoproliferative disorders, highlighting the critical role of T cells in achieving the EBV-host balance. Recent studies reveal that the EBV protein, LMP1, triggers robust T-cell responses against multiple tumor-associated antigens (TAAs) in B cells. Additionally, EBV-specific T cells have been identified in EBV-unrelated cancers, raising questions about their role in antitumor immunity. Herein, we summarize T-cell responses in EBV-related cancers, considering latency patterns, host immune status, and factors like human leukocyte antigen (HLA) susceptibility, which may affect immune outcomes. We discuss EBV-induced TAA-specific T cell responses and explore the potential roles of EBV-specific T cell subsets in tumor microenvironments. We also describe T-cell immunotherapy strategies that harness EBV antigens, ranging from EBV-specific T cells to T cell receptor-engineered T cells. Lastly, we discuss the involvement of γδ T-cells in EBV infection and associated diseases, aiming to elucidate the comprehensive interplay between EBV and T-cell immunity.
Collapse
Affiliation(s)
| | - Miao Xu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Sacco KA, Notarangelo LD, Delmonte OM. When to suspect inborn errors of immunity in Epstein-Barr virus-related lymphoproliferative disorders. Clin Microbiol Infect 2023; 29:457-462. [PMID: 36209991 PMCID: PMC10066820 DOI: 10.1016/j.cmi.2022.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/20/2022] [Accepted: 10/01/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND More than 95% of humans have been infected with Epstein-Barr virus (EBV) and develop anti-EBV IgG antibodies, conferring immunity. However, among specific populations, EBV may induce a range of B-cell lymphoproliferative disorders (LPDs). EBV may also contribute to T-cell and natural killer (NK)-cell lymphoproliferation. The immune system is essential to prevent infection and development of cancer. Inborn errors of immunity (IEIs) are a heterogenous group of more than 450 genetic disorders predisposing to severe and/or recurrent infection, autoimmunity, autoinflammation, or early-onset/severe neoplasia or lymphoproliferation. Monogenic disorders of T-cell and B-cell signalling are classic IEIs that predispose to EBV-associated LPDs. OBJECTIVES We aimed to outline the various clinical manifestations of EBV-associated LPDs and the underlying IEIs associated with such presentations and discuss the recommended management and therapeutic options pertaining to these disorders. SOURCES We searched PubMed, Embase, and Web of Science Core Collection on 30 September 2021. Clinical studies, systematic reviews, narrative reviews, and case reports were identified through search strategy and cross reference from primary literature. CONTENT Effective T-cell and NK-cell cytotoxicity towards EBV-infected B cells relies on intact MAGT1-dependent NKG2D pathways and signalling lymphocyte activation molecular-associated protein-dependent signalling lymphocyte activation molecular receptors. The interaction between CD27 and CD70 is also critical to drive the expansion of EBV-specific T cells. IEIs due to T-cell and B-cell signalling defects and/or impaired T-cell and NK-cell cytotoxicity predispose to EBV-related lymphoproliferation. This includes classic disorders such as X-linked lymphoproliferative disease 1 (due to SH2D1A mutations), X-linked lymphoproliferative disease 2 (XIAP), and other genetic diseases, such as ITK, MAGT1, CD27, CD70, CTPS1, RASGRP1, and CORO1A deficiencies. EBV-driven lymphoproliferation may manifest to a lesser degree in MST1/STK4, DOCK8, STIM1, CORO1A, IL21R, PIK3CD gain-of-function, and PI3KR1 deficiencies. IMPLICATIONS Early screening for IEIs is indicated in cases of EBV-related lymphoproliferation because different forms of IEIs have specific prognostic and therapeutic implications.
Collapse
Affiliation(s)
- Keith A Sacco
- Laboratory of Clinical Immunology and Microbiology, Immune Deficiency Genetics Section, National Institutes of Health, Bethesda, MD, USA
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, Immune Deficiency Genetics Section, National Institutes of Health, Bethesda, MD, USA
| | - Ottavia M Delmonte
- Laboratory of Clinical Immunology and Microbiology, Immune Deficiency Genetics Section, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
6
|
Eiz-Vesper B, Ravens S, Maecker-Kolhoff B. αβ and γδ T-cell responses to Epstein-Barr Virus: insights in immunocompetence, immune failure and therapeutic augmentation in transplant patients. Curr Opin Immunol 2023; 82:102305. [PMID: 36963323 DOI: 10.1016/j.coi.2023.102305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/19/2023] [Accepted: 02/21/2023] [Indexed: 03/26/2023]
Abstract
Epstein-Barr Virus (EBV) is a human gamma herpes virus, which causes several diseases in immunocompetent (mononucleosis, chronic fatigue syndrome, gastric cancer, endemic Burkitt's lymphoma, head and neck cancer) and immunosuppressed (post-transplant lymphoproliferative disease, EBV-associated soft tissue tumors) patients. It elicits a complex humoral and cellular immune response with both innate and adaptive immune components. Substantial progress has been made in understanding the interplay of immune cells in EBV-associated diseases in recent years, and several therapeutic approaches have been developed to augment cellular immunity toward EBV for control of EBV-associated malignancy. This review will focus on recent developments in immunosuppressed transplant recipients.
Collapse
Affiliation(s)
- Britta Eiz-Vesper
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Germany; CRC900 Microbial persistence and its control; German Center for Infection Research (DZIF)
| | - Sarina Ravens
- CRC900 Microbial persistence and its control; Institute of Immunology, Hannover Medical School, Germany
| | - Britta Maecker-Kolhoff
- CRC900 Microbial persistence and its control; German Center for Infection Research (DZIF); Department of Pediatric Hematology and Oncology, Hannover Medical School, Germany.
| |
Collapse
|
7
|
Desimio MG, Covino DA, Rivalta B, Cancrini C, Doria M. The Role of NK Cells in EBV Infection and Related Diseases: Current Understanding and Hints for Novel Therapies. Cancers (Basel) 2023; 15:cancers15061914. [PMID: 36980798 PMCID: PMC10047181 DOI: 10.3390/cancers15061914] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
The Epstein-Barr virus (EBV) is a ubiquitous herpesvirus most often transmitted during infancy and infecting the vast majority of human beings. Usually, EBV infection is nearly asymptomatic and results in life-long persistency of the virus in a latent state under the control of the host immune system. Yet EBV can cause an acute infectious mononucleosis (IM), particularly in adolescents, and is associated with several malignancies and severe diseases that pose a serious threat to individuals with specific inborn error of immunity (IEI). While there is a general consensus on the requirement for functional CD8 T cells to control EBV infection, the role of the natural killer (NK) cells of the innate arm of immunity is more enigmatic. Here we provide an overview of the interaction between EBV and NK cells in the immunocompetent host as well as in the context of primary and secondary immunodeficiencies. Moreover, we report in vitro data on the mechanisms that regulate the capacity of NK cells to recognize and kill EBV-infected cell targets and discuss the potential of recently optimized NK cell-based immunotherapies for the treatment of EBV-associated diseases.
Collapse
Affiliation(s)
- Maria G Desimio
- Primary Immunodeficiency Research Unit, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Daniela A Covino
- Primary Immunodeficiency Research Unit, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Beatrice Rivalta
- Primary Immunodeficiency Research Unit, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Caterina Cancrini
- Primary Immunodeficiency Research Unit, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Margherita Doria
- Primary Immunodeficiency Research Unit, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| |
Collapse
|
8
|
Kinzel M, Kalra A, Tripathi G, Dharmani-Khan P, Khan F, Storek J. Lack of both donor and recipient anti-EBV T cells in EBV seronegative recipients of grafts from seropositive donors. Bone Marrow Transplant 2023; 58:332-333. [PMID: 36450829 DOI: 10.1038/s41409-022-01884-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 12/03/2022]
Affiliation(s)
- Megan Kinzel
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| | - Amit Kalra
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Gaurav Tripathi
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Poonam Dharmani-Khan
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Alberta Precision Labs, Calgary, AB, Canada
| | - Faisal Khan
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Alberta Precision Labs, Calgary, AB, Canada
| | - Jan Storek
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
9
|
Damania B, Kenney SC, Raab-Traub N. Epstein-Barr virus: Biology and clinical disease. Cell 2022; 185:3652-3670. [PMID: 36113467 PMCID: PMC9529843 DOI: 10.1016/j.cell.2022.08.026] [Citation(s) in RCA: 125] [Impact Index Per Article: 62.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 01/26/2023]
Abstract
Epstein-Barr virus (EBV) is a ubiquitous, oncogenic virus that is associated with a number of different human malignancies as well as autoimmune disorders. The expression of EBV viral proteins and non-coding RNAs contribute to EBV-mediated disease pathologies. The virus establishes life-long latency in the human host and is adept at evading host innate and adaptive immune responses. In this review, we discuss the life cycle of EBV, the various functions of EBV-encoded proteins and RNAs, the ability of the virus to activate and evade immune responses, as well as the neoplastic and autoimmune diseases that are associated with EBV infection in the human population.
Collapse
Affiliation(s)
- Blossom Damania
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Shannon C Kenney
- Department of Oncology, McArdle Laboratory for Cancer Research, and Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Nancy Raab-Traub
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
10
|
Gay L, Mezouar S, Cano C, Frohna P, Madakamutil L, Mège JL, Olive D. Role of Vγ9vδ2 T lymphocytes in infectious diseases. Front Immunol 2022; 13:928441. [PMID: 35924233 PMCID: PMC9340263 DOI: 10.3389/fimmu.2022.928441] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/27/2022] [Indexed: 12/22/2022] Open
Abstract
The T cell receptor Vγ9Vδ2 T cells bridge innate and adaptive antimicrobial immunity in primates. These Vγ9Vδ2 T cells respond to phosphoantigens (pAgs) present in microbial or eukaryotic cells in a butyrophilin 3A1 (BTN3) and butyrophilin 2A1 (BTN2A1) dependent manner. In humans, the rapid expansion of circulating Vγ9Vδ2 T lymphocytes during several infections as well as their localization at the site of active disease demonstrates their important role in the immune response to infection. However, Vγ9Vδ2 T cell deficiencies have been observed in some infectious diseases such as active tuberculosis and chronic viral infections. In this review, we are providing an overview of the mechanisms of Vγ9Vδ2 T cell-mediated antimicrobial immunity. These cells kill infected cells mainly by releasing lytic mediators and pro-inflammatory cytokines and inducing target cell apoptosis. In addition, the release of chemokines and cytokines allows the recruitment and activation of immune cells, promoting the initiation of the adaptive immune response. Finaly, we also describe potential new therapeutic tools of Vγ9Vδ2 T cell-based immunotherapy that could be applied to emerging infections.
Collapse
Affiliation(s)
- Laetitia Gay
- Aix-Marseille Univ, Intitut Recherche pour le Développement (IRT), Assistance Publique Hôpitaux de Marseille (APHM), Microbe, Evolution, Phylogeny, Infection (MEPHI), Marseille, France
- Immunology Department, IHU-Méditerranée Infection, Marseille, France
- ImCheck Therapeutics, Marseille, France
| | - Soraya Mezouar
- Aix-Marseille Univ, Intitut Recherche pour le Développement (IRT), Assistance Publique Hôpitaux de Marseille (APHM), Microbe, Evolution, Phylogeny, Infection (MEPHI), Marseille, France
- Immunology Department, IHU-Méditerranée Infection, Marseille, France
| | | | | | | | - Jean-Louis Mège
- Aix-Marseille Univ, Intitut Recherche pour le Développement (IRT), Assistance Publique Hôpitaux de Marseille (APHM), Microbe, Evolution, Phylogeny, Infection (MEPHI), Marseille, France
- Immunology Department, IHU-Méditerranée Infection, Marseille, France
- Aix-Marseille Univ, APHM, Hôpital de la Conception, Laboratoire d’Immunologie, Marseille, France
| | - Daniel Olive
- Centre pour la Recherche sur le Cancer de Marseille (CRCM), Inserm UMR1068, Centre national de la recherche scientifique (CNRS) UMR7258, Institut Paoli Calmettes, Marseille, France
| |
Collapse
|
11
|
Gay L, Mezouar S, Cano C, Foucher E, Gabriac M, Fullana M, Madakamutil L, Mège JL, Olive D. BTN3A Targeting Vγ9Vδ2 T Cells Antimicrobial Activity Against Coxiella burnetii-Infected Cells. Front Immunol 2022; 13:915244. [PMID: 35833118 PMCID: PMC9272908 DOI: 10.3389/fimmu.2022.915244] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/30/2022] [Indexed: 12/26/2022] Open
Abstract
Vγ9Vδ2 T cells have been reported to participate to the immune response against infectious diseases such as the Q fever caused by Coxiella burnetii infection. Indeed, the number and proportion of Vγ9Vδ2 T cells are increased during the acute phase of Q fever. Human Vγ9Vδ2 T cell responses are triggered by phosphoantigens (pAgs) produced by pathogens and malignant cells, that are sensed via the membrane receptors butyrophilin-3A1 (BTN3A1) and -2A1 (BTN2A1). Here, by using CRISPR-Cas9 inactivation in THP-1 cells, we show that BTN3A and BTN2A are required to Vγ9Vδ2 T cell response to C. burnetii infection, though not directly involved in the infection process. Furthermore, C. burnetii-infected monocytes display increased BTN3A and BTN2A expression and induce Vγ9Vδ2 T cell activation that can be inhibited by specific antagonist mAb. More importantly, we show that the antimicrobial functions of Vγ9Vδ2 T cells towards C. burnetii are enhanced in the presence of an BTN3A activating antibody. This supports the role of Vγ9Vδ2 T cells in the control of C. burnetii infection and argues in favor of targeting these cells as an alternative treatment strategy for infectious diseases caused by intracellular bacteria.
Collapse
Affiliation(s)
- Laetitia Gay
- Aix-Marseille University (Univ), IRD, Assistance Publique Hopitaux de Marseille (APHM), Microbe, Evolution, Phylogeny, Infection (MEPHI), Marseille, France
- IHU-Méditerranée Infection, Marseille, France
- ImCheck Therapeutics, Marseille, France
| | - Soraya Mezouar
- Aix-Marseille University (Univ), IRD, Assistance Publique Hopitaux de Marseille (APHM), Microbe, Evolution, Phylogeny, Infection (MEPHI), Marseille, France
- IHU-Méditerranée Infection, Marseille, France
| | | | | | | | | | | | - Jean-Louis Mège
- Aix-Marseille University (Univ), IRD, Assistance Publique Hopitaux de Marseille (APHM), Microbe, Evolution, Phylogeny, Infection (MEPHI), Marseille, France
- IHU-Méditerranée Infection, Marseille, France
- Aix-Marseille University (Univ), Assistance Publique Hopitaux de Marseille (APHM), Hôpital de la Conception, Laboratoire d’Immunologie, Marseille, France
| | - Daniel Olive
- Centre de Recheche contre le cancer de Marseille (CRCM), Inserm UMR1068, Centre national de la recherche scientifique (CNRS) UMR7258, Institut Paoli Calmettes, Marseille, France
- *Correspondence: Daniel Olive,
| |
Collapse
|
12
|
Walwyn-Brown K, Pugh J, Cocker AT, Beyzaie N, Singer BB, Olive D, Guethlein LA, Parham P, Djaoud Z. Phosphoantigen-stimulated γδ T cells suppress natural killer cell-responses to missing-self. Cancer Immunol Res 2022; 10:558-570. [PMID: 35263761 DOI: 10.1158/2326-6066.cir-21-0696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 01/14/2022] [Accepted: 03/04/2022] [Indexed: 11/16/2022]
Abstract
γδ T cells stimulated by phosphoantigens (pAg) are potent effectors that secrete Th1 cytokines and kill tumor cells. Consequently, they are considered candidates for use in cancer immunotherapy. However, they have proven only moderately effective in several clinical trials. We studied the consequences of pAg-stimulated γδ T-cell interactions with Natural Killer (NK) cells and CD8+ T cells, major innate and adaptive effectors, respectively. We found that pAg-stimulated γδ T cells suppressed NK-cell responses to "missing-self" but had no effect on antigen-specific CD8+ T-cell responses. Extensive analysis of the secreted cytokines showed that pAg-stimulated γδ T cells had a pro-inflammatory profile. CMV-pp65-specific CD8+ T cells primed with pAg-stimulated γδ T cells showed little effect on responses to pp65-loaded target cells. By contrast, NK cells primed similarly with γδ T cells had impaired capacity to degranulate and produce IFNγ in response to HLA class I-deficient targets. This effect depended on BTN3A1 and required direct contact between NK cells and γδ T cells. γδ T cell-priming of NK cells also led to a downregulation of NKG2D and NKp44 on NK cells. Every NK-cell subset was affected by γδ T cell-mediated immunosuppression, but the strongest effect was on KIR+NKG2A- NK cells. We therefore report a previously unknown function for γδ T cells, as brakes of NK-cell responses to "missing-self". This provides a new perspective for optimizing the use of γδ T cells in cancer immunotherapy and for assessing their role in immune responses to pAg-producing pathogens.
Collapse
Affiliation(s)
| | | | | | | | | | - Daniel Olive
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, CRCM,, Marseille, France
| | | | | | | |
Collapse
|
13
|
Jiang P, Nolte IM, Hepkema BG, Stulp M, van den Berg A, Diepstra A. Killer Cell Immunoglobulin-Like Receptor Haplotype B Modulates Susceptibility to EBV-Associated Classic Hodgkin Lymphoma. Front Immunol 2022; 13:829943. [PMID: 35154153 PMCID: PMC8828906 DOI: 10.3389/fimmu.2022.829943] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 01/07/2022] [Indexed: 11/21/2022] Open
Abstract
Tumor cells of classic Hodgkin lymphoma (cHL) are derived from antigen presenting B cells that are infected by Epstein Barr virus (EBV) in ~30% of patients. Polymorphic Killer cell immunoglobulin-like receptors (KIRs) expressed on NK cells interact with human leukocyte antigen (HLA) class I and play a key role in immune surveillance against virally infected cells and tumor cells. We investigated the effect of KIR types on cHL susceptibility overall (n=211) and in EBV-stratified subgroups using the Dutch GoNL cohort as controls (n=498). The frequency of the KIR haplotype B subgroup was significantly different between EBV+ and EBV− cHL patients (62% vs. 77%, p=0.04) and this difference was more pronounced in nodular sclerosis (NS) cHL (49% vs. 79%, p=0.0003). The frequency of KIR haplotype B subgroup was significantly lower in EBV+ NS cHL compared to controls (49% vs. 67%, p=0.01). Analyses of known KIR – HLA interaction pairs revealed lower carrier frequencies of KIR2DS2 – HLA-C1 (29% vs. 46%, p=0.03) and KIR2DL2 – HLA-C1 (29% vs. 45%, p=0.04) in EBV+ NS cHL patients compared to controls. Carriers of the KIR haplotype B subgroup are less likely to develop EBV+ NS cHL, probably because of a more efficient control over EBV-infected B cells.
Collapse
Affiliation(s)
- Peijia Jiang
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Ilja M Nolte
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Bouke G Hepkema
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Marijke Stulp
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Anke van den Berg
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Arjan Diepstra
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
14
|
The Role of γδ T Cells as a Line of Defense in Viral Infections after Allogeneic Stem Cell Transplantation: Opportunities and Challenges. Viruses 2022; 14:v14010117. [PMID: 35062321 PMCID: PMC8779492 DOI: 10.3390/v14010117] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/30/2021] [Accepted: 01/05/2022] [Indexed: 02/04/2023] Open
Abstract
In the complex interplay between inflammation and graft-versus-host disease (GVHD) after allogeneic stem cell transplantation (allo-HSCT), viral reactivations are often observed and cause substantial morbidity and mortality. As toxicity after allo-HSCT within the context of viral reactivations is mainly driven by αβ T cells, we describe that by delaying αβ T cell reconstitution through defined transplantation techniques, we can harvest the full potential of early reconstituting γδ T cells to control viral reactivations. We summarize evidence of how the γδ T cell repertoire is shaped by CMV and EBV reactivations after allo-HSCT, and their potential role in controlling the most important, but not all, viral reactivations. As most γδ T cells recognize their targets in an MHC-independent manner, γδ T cells not only have the potential to control viral reactivations but also to impact the underlying hematological malignancies. We also highlight the recently re-discovered ability to recognize classical HLA-molecules through a γδ T cell receptor, which also surprisingly do not associate with GVHD. Finally, we discuss the therapeutic potential of γδ T cells and their receptors within and outside the context of allo-HSCT, as well as the opportunities and challenges for developers and for payers.
Collapse
|
15
|
Münz C. Co-Stimulatory Molecules during Immune Control of Epstein Barr Virus Infection. Biomolecules 2021; 12:biom12010038. [PMID: 35053187 PMCID: PMC8774114 DOI: 10.3390/biom12010038] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 01/17/2023] Open
Abstract
The Epstein Barr virus (EBV) is one of the prominent human tumor viruses, and it is efficiently immune-controlled in most virus carriers. Cytotoxic lymphocytes strongly expand during symptomatic primary EBV infection and in preclinical in vivo models of this tumor virus infection. In these models and patients with primary immunodeficiencies, antibody blockade or deficiencies in certain molecular pathways lead to EBV-associated pathologies. In addition to T, NK, and NKT cell development, as well as their cytotoxic machinery, a set of co-stimulatory and co-inhibitory molecules was found to be required for EBV-specific immune control. The role of CD27/CD70, 4-1BB, SLAMs, NKG2D, CD16A/CD2, CTLA-4, and PD-1 will be discussed in this review. Some of these have just been recently identified as crucial for EBV-specific immune control, and for others, their important functions during protection were characterized in in vivo models of EBV infection and its immune control. These insights into the phenotype of cytotoxic lymphocytes that mediate the near-perfect immune control of EBV-associated malignancies might also guide immunotherapies against other tumors in the future.
Collapse
Affiliation(s)
- Christian Münz
- Department of Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, 8057 Zurich, Switzerland
| |
Collapse
|
16
|
Martini F, Champagne E. The Contribution of Human Herpes Viruses to γδ T Cell Mobilisation in Co-Infections. Viruses 2021; 13:v13122372. [PMID: 34960641 PMCID: PMC8704314 DOI: 10.3390/v13122372] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022] Open
Abstract
γδ T cells are activated in viral, bacterial and parasitic infections. Among viruses that promote γδ T cell mobilisation in humans, herpes viruses (HHVs) occupy a particular place since they infect the majority of the human population and persist indefinitely in the organism in a latent state. Thus, other infections should, in most instances, be considered co-infections, and the reactivation of HHV is a serious confounding factor in attributing γδ T cell alterations to a particular pathogen in human diseases. We review here the literature data on γδ T cell mobilisation in HHV infections and co-infections, and discuss the possible contribution of HHVs to γδ alterations observed in various infectious settings. As multiple infections seemingly mobilise overlapping γδ subsets, we also address the concept of possible cross-protection.
Collapse
|
17
|
Feng M, Duan R, Gao Y, Zhang H, Qiao Y, Li Q, Zhao F. Role of Epstein-Barr Virus and Human Papillomavirus Coinfection in Cervical Intraepithelial Neoplasia in Chinese Women Living With HIV. Front Cell Infect Microbiol 2021; 11:703259. [PMID: 34557425 PMCID: PMC8453025 DOI: 10.3389/fcimb.2021.703259] [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: 04/30/2021] [Accepted: 08/19/2021] [Indexed: 01/04/2023] Open
Abstract
Given that only a small percentage of human papillomavirus (HPV)-positive women develop cancer, HPV is necessary but insufficient for carcinogenesis. Mucosally transmitted viral cofactors appear to contribute to HPV-related cervical cancer, such as Epstein-Barr virus (EBV), but previous studies have shown inconsistent outcomes. The exact role of EBV in cervical cancer remains unclear, and more studies are needed to determine its involvement. In this study, we describe the prevalence of EBV and HPV coinfection in HIV-positive women and explore how abnormal host immune status induced by viral coinfections modulates epithelial gene expression. We found a significant correlation between EBV-HPV coinfection and the incidence of high-grade cervical intraepithelial neoplasia (CIN2+). RNA sequencing indicated that CIN tissues coinfected with EBV and HPV led to significant changes in the gene expression of epithelial differentiation and development compared to normal tissues with HPV infection alone. In particular, several differentially expressed genes (DEGs) are closely associated with cancer, such as CACNG4, which was confirmed to be upregulated at both the mRNA and protein levels. Therefore, these findings provide some evidence that EBV may act as a cofactor or mediator in HPV-related cervical cancer. Specific genes or proteins, such as CACNG4, may serve as biomarkers that can risk stratify patients based on pathological changes in the cervix.
Collapse
Affiliation(s)
- Min Feng
- Institute of Medical Biology, Chinese Academy of Medicine Sciences and Peking Union Medical College, Kunming, China
| | - Rufei Duan
- Department of Gynecology, The Third Affiliated Hospital of Kunming Medical University/Yunnan Cancer Hospital/Yunnan Cancer Center, Kunming, China.,Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yang Gao
- Institute of Medical Biology, Chinese Academy of Medicine Sciences and Peking Union Medical College, Kunming, China
| | - Han Zhang
- Institute of Medical Biology, Chinese Academy of Medicine Sciences and Peking Union Medical College, Kunming, China
| | - Youlin Qiao
- Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qihan Li
- Institute of Medical Biology, Chinese Academy of Medicine Sciences and Peking Union Medical College, Kunming, China
| | - Fanghui Zhao
- Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
18
|
Morrison AL, Sharpe S, White AD, Bodman-Smith M. Cheap and Commonplace: Making the Case for BCG and γδ T Cells in COVID-19. Front Immunol 2021; 12:743924. [PMID: 34567010 PMCID: PMC8455994 DOI: 10.3389/fimmu.2021.743924] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 08/19/2021] [Indexed: 12/26/2022] Open
Abstract
Antigen-specific vaccines developed for the COVID-19 pandemic demonstrate a remarkable achievement and are currently being used in high income countries with much success. However, new SARS-CoV-2 variants are threatening this success via mutations that lessen the efficacy of antigen-specific antibodies. One simple approach to assisting with this issue is focusing on strategies that build on the non-specific protection afforded by the innate immune response. The BCG vaccine has been shown to provide broad protection beyond tuberculosis disease, including against respiratory viruses, and ongoing studies are investigating its efficacy as a tool against SARS-CoV-2. Gamma delta (γδ) T cells, particularly the Vδ2 subtype, undergo rapid expansion after BCG vaccination due to MHC-independent mechanisms. Consequently, γδ T cells can produce diverse defenses against virally infected cells, including direct cytotoxicity, death receptor ligands, and pro-inflammatory cytokines. They can also assist in stimulating the adaptive immune system. BCG is affordable, commonplace and non-specific, and therefore could be a useful tool to initiate innate protection against new SARS-CoV-2 variants. However, considerations must also be made to BCG vaccine supply and the prioritization of countries where it is most needed to combat tuberculosis first and foremost.
Collapse
Affiliation(s)
| | - Sally Sharpe
- Public Health England, National Infection Service, Porton Down, United Kingdom
| | - Andrew D. White
- Public Health England, National Infection Service, Porton Down, United Kingdom
| | - Mark Bodman-Smith
- Infection and Immunity Research Institute, St George’s University of London, London, United Kingdom
| |
Collapse
|
19
|
Liu J, Gao H, Xu LP, Mo XD, Liu R, Liang S, Wu N, Wang M, Wang Z, Chang YJ, Wang Y, Zhang XH, Huang XJ. Immunosuppressant indulges EBV reactivation and related lymphoproliferative disease by inhibiting Vδ2 + T cells activities after hematopoietic transplantation for blood malignancies. J Immunother Cancer 2021; 8:jitc-2019-000208. [PMID: 32221014 PMCID: PMC7206968 DOI: 10.1136/jitc-2019-000208] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2020] [Indexed: 12/16/2022] Open
Abstract
Background Following the extensive use of immunosuppressive drugs in the clinic, immunosuppression-associated side effects have received increasing attention. Epstein-Barr virus (EBV) reactivation and related lymphoproliferative diseases (LPD) are the lethal complications observed after allogeneic hematopoietic cell transplantation (alloHCT). While studies generally suggest an association between immunosuppressants and EBV reactivation, the effects of specific immunosuppressive drugs and which T-cell subsets mediate these correlations are unclear. Vδ2+ T cells are correlated with EBV reactivation after alloHCT. Researchers have not determined whether Vδ2+ T-cell activities are affected by immunosuppressants and thereby facilitate EBV reactivation and related LPD. Methods A clinical cohort study of 170 patients with hematopoietic malignancies who received haploidentical hematopoietic cell transplantation (haploHCT) was performed to investigate whether the early cessation of mycophenolate mofetil (MMF) decreases EBV reactivation and related LPD and to determine whether this change is associated with the recovery of Vδ2 + T cells after transplantation. The effects of MMF on the expansion and anti-EBV capacity of Vδ2+ T cells were detected in vitro and in an immunodeficient mouse model. Results A reduction in the course of MMF significantly improved the recovery of Vδ2+ T cells from 30 to 90 days after haploHCT (p=0.002, p=0.042 and p=0.035, respectively), accompanied by a significant decrease in EBV reactivation (from 26% to 13%, p=0.033) and EBV-LPD (from 10.6% to 2.4%, p=0.029). The day-30 Vδ2+ T level remained an independent factor for EBV reactivation in patients with different MMF durations (p=0.007). In the in-vitro experiments, MMF inhibited Vδ2+ T-cell expansion and its cytotoxicity on EBV-transformed malignant cells. Furthermore, the therapeutic and prophylactic effects of adoptively transferred human Vδ2+ T cells were attenuated by the MMF treatment in immunodeficient mice with EBV-LPD. Conclusions These results elucidated a negative effect of immunosuppressants on the anti-EBV capacity of Vδ2+ T cells. Strategies that appropriately relieve the immunosuppression may improve anti-EBV immunity by increasing the activity of Vδ2+ T cells after alloHCT.
Collapse
Affiliation(s)
- Jiangying Liu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Haitao Gao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Lan-Ping Xu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Dong Mo
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Ruoyang Liu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Shuang Liang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Ning Wu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ming Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhidong Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Ying-Jun Chang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yu Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China .,Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.,Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
20
|
Ye W, Kong X, Zhang W, Weng Z, Wu X. The Roles of γδ T Cells in Hematopoietic Stem Cell Transplantation. Cell Transplant 2021; 29:963689720966980. [PMID: 33073597 PMCID: PMC7784584 DOI: 10.1177/0963689720966980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The αβ T-cell-depleted hematopoietic stem cell transplantation (HSCT) leads to lower relapse and better outcome, and may correlate strongly with expansion of donor-derived γδ T cells. γδ T cells play an important role in immune reconstitution and can exert a graft-versus-leukemia effect after HSCT. This review showed the recent literature on immune functions of γδ T cells after HSCT. The discrepancies between studies of γδ T cells in graft-versus-host disease may cause by its heterogeneous and various distinct subsets. And reconstitution of γδ T cells may play a potential immunoregulatory role in the infections after HSCT.
Collapse
Affiliation(s)
- Wanyi Ye
- Institute of Hematology, School of Medicine, 47885Jinan University, Guangzhou, China
| | - Xueting Kong
- Institute of Hematology, School of Medicine, 47885Jinan University, Guangzhou, China
| | - Wenbin Zhang
- Institute of Hematology, School of Medicine, 47885Jinan University, Guangzhou, China
| | - Zheng Weng
- Institute of Hematology, School of Medicine, 47885Jinan University, Guangzhou, China
| | - Xiuli Wu
- Institute of Hematology, School of Medicine, 47885Jinan University, Guangzhou, China.,Key Laboratory for Regenerative Medicine of Ministry of Education, 47885Jinan University, Guangzhou, China
| |
Collapse
|
21
|
Münz C. Immune Escape by Non-coding RNAs of the Epstein Barr Virus. Front Microbiol 2021; 12:657387. [PMID: 34234755 PMCID: PMC8257079 DOI: 10.3389/fmicb.2021.657387] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 05/28/2021] [Indexed: 01/20/2023] Open
Abstract
Epstein Barr virus (EBV) is one of the most successful pathogens of humans, persistently colonizing more than 95% of the adult human population. At the same time EBV encodes oncogenes that can readily transform human B cells in culture and threaten healthy virus carriers with lymphomagenesis. Cytotoxic lymphocytes have been identified in experimental models and by primary immunodeficiencies as the main protective immune compartments controlling EBV. EBV has reached a stalemate with these cytotoxic T and innate lymphocytes to ensure persistence in most infected humans. Recent evidence suggests that the non-coding RNAs of the virus contribute to viral immune escape to prevent immune eradication. This knowledge might be used in the future to attenuate EBV for vaccine development against this human tumor virus that was discovered more than 55 years ago.
Collapse
Affiliation(s)
- Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
22
|
Abstract
In all human cells, human leukocyte antigen (HLA) class I glycoproteins assemble with a peptide and take it to the cell surface for surveillance by lymphocytes. These include natural killer (NK) cells and γδ T cells of innate immunity and αβ T cells of adaptive immunity. In healthy cells, the presented peptides derive from human proteins, to which lymphocytes are tolerant. In pathogen-infected cells, HLA class I expression is perturbed. Reduced HLA class I expression is detected by KIR and CD94:NKG2A receptors of NK cells. Almost any change in peptide presentation can be detected by αβ CD8+ T cells. In responding to extracellular pathogens, HLA class II glycoproteins, expressed by specialized antigen-presenting cells, present peptides to αβ CD4+ T cells. In comparison to the families of major histocompatibility complex (MHC) class I, MHC class II and αβ T cell receptors, the antigenic specificity of the γδ T cell receptors is incompletely understood.
Collapse
Affiliation(s)
- Zakia Djaoud
- Department of Structural Biology and Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, USA; ,
| | - Peter Parham
- Department of Structural Biology and Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, USA; ,
| |
Collapse
|
23
|
Fournier B, Latour S. Immunity to EBV as revealed by immunedeficiencies. Curr Opin Immunol 2021; 72:107-115. [PMID: 33989894 DOI: 10.1016/j.coi.2021.04.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/29/2021] [Accepted: 04/05/2021] [Indexed: 02/06/2023]
Abstract
Epstein-Barr virus infection is the most common viral latent infection in humans and represents one prototypical model to study immunity to viral infections. In that respect, inborn errors of immunity (IEIs) or primary immunodeficiencies (PIDs) predisposing to severe and chronic EBV infections provide peculiar examples to decipher-specific molecular and cellular components involved in the immune control of EBV-infected cells. Herein, we discuss the recent knowledge and concepts arising from these studies, with a particular focus on 'atypical' EBV infections when EBV enters T, NK and smooth muscle cells, instead of the common 'typical' infection of B cells.
Collapse
Affiliation(s)
- Benjamin Fournier
- Laboratory of Lymphocyte Activation and Susceptibility to EBV Infection, INSERM UMR 1163, Imagine Institute, Paris, France; Université de Paris, F75006 Paris, France; Department of Pediatric Immunology, Hematology and Rheumatology, Necker-Enfants Malades, Paris, France
| | - Sylvain Latour
- Laboratory of Lymphocyte Activation and Susceptibility to EBV Infection, INSERM UMR 1163, Imagine Institute, Paris, France; Université de Paris, F75006 Paris, France.
| |
Collapse
|
24
|
Deng Y, Münz C. Roles of Lytic Viral Replication and Co-Infections in the Oncogenesis and Immune Control of the Epstein-Barr Virus. Cancers (Basel) 2021; 13:2275. [PMID: 34068598 PMCID: PMC8126045 DOI: 10.3390/cancers13092275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 11/16/2022] Open
Abstract
Epstein-Barr virus (EBV) is the prototypic human tumor virus whose continuous lifelong immune control is required to prevent lymphomagenesis in the more than 90% of the human adult population that are healthy carriers of the virus. Here, we review recent evidence that this immune control has not only to target latent oncogenes, but also lytic replication of EBV. Furthermore, genetic variations identify the molecular machinery of cytotoxic lymphocytes as essential for this immune control and recent studies in mice with reconstituted human immune system components (humanized mice) have begun to provide insights into the mechanistic role of these molecules during EBV infection. Finally, EBV often does not act in isolation to cause disease. Some of EBV infection-modulating co-infections, including human immunodeficiency virus (HIV) and Kaposi sarcoma-associated herpesvirus (KSHV), have been modeled in humanized mice. These preclinical in vivo models for EBV infection, lymphomagenesis, and cell-mediated immune control do not only promise a better understanding of the biology of this human tumor virus, but also the possibility to explore vaccine candidates against it.
Collapse
Affiliation(s)
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, 8057 Zürich, Switzerland;
| |
Collapse
|
25
|
Natural Killer Cells in Post-Transplant Lymphoproliferative Disorders. Cancers (Basel) 2021; 13:cancers13081836. [PMID: 33921413 PMCID: PMC8068932 DOI: 10.3390/cancers13081836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/19/2022] Open
Abstract
Post-transplant lymphoproliferative disorders (PTLDs) are life-threatening complications arising after solid organ or hematopoietic stem cell transplantations. Although the majority of these lymphoproliferations are of B cell origin, and are frequently associated with primary Epstein-Barr virus (EBV) infection or reactivation in the post-transplant period, rare cases of T cell and natural killer (NK) cell-originated PTLDs have also been described. A general assumption is that PTLDs result from the impairment of anti-viral and anti-tumoral immunosurveillance due to the long-term use of immunosuppressants in transplant recipients. T cell impairment is known to play a critical role in the immune-pathogenesis of post-transplant EBV-linked complications, while the role of NK cells has been less investigated, and is probably different between EBV-positive and EBV-negative PTLDs. As a part of the innate immune response, NK cells are critical for protecting hosts during the early response to virus-induced tumors. The complexity of their function is modulated by a myriad of activating and inhibitory receptors expressed on cell surfaces. This review outlines our current understanding of NK cells in the pathogenesis of PTLD, and discusses their potential implications for current PTLD therapies and novel NK cell-based therapies for the containment of these disorders.
Collapse
|
26
|
Caron J, Ridgley LA, Bodman-Smith M. How to Train Your Dragon: Harnessing Gamma Delta T Cells Antiviral Functions and Trained Immunity in a Pandemic Era. Front Immunol 2021; 12:666983. [PMID: 33854516 PMCID: PMC8039298 DOI: 10.3389/fimmu.2021.666983] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 03/12/2021] [Indexed: 12/23/2022] Open
Abstract
The emergence of viruses with pandemic potential such as the SARS-CoV-2 coronavirus causing COVID-19 poses a global health challenge. There is remarkable progress in vaccine technology in response to this threat, but their design often overlooks the innate arm of immunity. Gamma Delta (γδ) T cells are a subset of T cells with unique features that gives them a key role in the innate immune response to a variety of homeostatic alterations, from cancer to microbial infections. In the context of viral infection, a growing body of evidence shows that γδ T cells are particularly equipped for early virus detection, which triggers their subsequent activation, expansion and the fast deployment of antiviral functions such as direct cytotoxic pathways, secretion of cytokines, recruitment and activation of other immune cells and mobilization of a trained immunity memory program. As such, γδ T cells represent an attractive target to stimulate for a rapid and effective resolution of viral infections. Here, we review the known aspects of γδ T cells that make them crucial component of the immune response to viruses, and the ways that their antiviral potential can be harnessed to prevent or treat viral infection.
Collapse
Affiliation(s)
- Jonathan Caron
- Infection and Immunity Research Institute, St. George's University of London, London, United Kingdom
| | - Laura Alice Ridgley
- Infection and Immunity Research Institute, St. George's University of London, London, United Kingdom
| | - Mark Bodman-Smith
- Infection and Immunity Research Institute, St. George's University of London, London, United Kingdom
| |
Collapse
|
27
|
Innate and Adaptive Immune Correlates of Chronic and Self-limiting EBV DNAemia in Solid-organ Transplant Recipients. Transplantation 2021; 104:2373-2382. [PMID: 31985732 DOI: 10.1097/tp.0000000000003130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Epstein-Barr virus (EBV) DNAemia is a major risk factor for posttransplant lymphoproliferative disorder; however, immune correlates of EBV DNAemia in the transplant setting are limited. METHODS Peripheral blood mononuclear cells were collected from 30 transplant recipients with self-limiting EBV DNAemia (SLD; n = 11) or chronic EBV DNAemia (CD; n = 19) at enrollment and 4-8 weeks later. Mass cytometry was used to characterize innate and T-cell immune correlates of EBV DNAemia. Furthermore, flow cytometry was used to measure the frequency of EBV-specific T-cell responses between groups following stimulation with an EBV-infected cell lysate. RESULTS Unsupervised analysis of the innate compartment (CD3CD19 cells) identified 5 CD11c clusters at higher abundance in the SLD group (false discovery rate ≤ 1%). These clusters expressed CD11b, CD45RO, CD14, CD123, CD127, and CD38, among others. Unsupervised profiling of the T-cell compartment (CD3CD19) revealed 2 CD4 T-cell clusters at higher frequency among those with SLD (false discovery rate ≤ 1%), which expressed CD45RA, CCR7, CD27, CD28, and CD40L-suggestive of a naive T cell (TN). Manual biaxial gating confirmed increased frequencies of conventional dendritic cells (3.1% versus 2.1%; P = 0.023) and CD4 TN (4.4% versus 1.9%; P = 0.018) among those with SLD. Last, frequencies of interferon-γ-producing EBV-specific CD4 T cells were significantly lower in the CD group relative to those with SLD (4243 versus 250 cells/10 cells; P = 0.015). CONCLUSIONS CD is associated with a reduction of CD11c cells, CD4 TN, and interferon-γ-producing EBV-specific CD4 T cells, suggesting an interplay between innate and adaptive immune compartments may be important for regulating EBV DNAemia.
Collapse
|
28
|
Mbiribindi B, Pena JK, Arvedson MP, Moreno Romero C, McCarthy SR, Hatton OL, Esquivel CO, Martinez OM, Krams SM. Epstein-Barr virus peptides derived from latent cycle proteins alter NKG2A + NK cell effector function. Sci Rep 2020; 10:19973. [PMID: 33203899 PMCID: PMC7673117 DOI: 10.1038/s41598-020-76344-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022] Open
Abstract
Natural killer (NK) cells control viral infection through the interaction between inhibitory receptors and human leukocyte antigen (HLA) ligands and bound peptide. NK cells expressing the inhibitory receptor NKG2A/CD94 recognize and respond to autologous B cells latently infected with Epstein-Barr virus (EBV). The mechanism is not yet understood, thus we investigated peptides derived from seven latent proteins of EBV in the interaction of NKG2A and its ligand HLA-E. Functional analysis demonstrated that EBV peptides can bind to HLA-E and block inhibition of NK cell effector function. Moreover, analysis of DNA from 79 subjects showed sequence variations in the latent protein, LMP1, which alters NK responses to EBV. We provide evidence that peptides derived from EBV latent cycle proteins can impair the recognition of NKG2A despite being presented by HLA-E, resulting in NK cell activation.
Collapse
Affiliation(s)
- Berenice Mbiribindi
- Division of Abdominal Transplantation, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Josselyn K Pena
- Division of Abdominal Transplantation, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Matthew P Arvedson
- Division of Abdominal Transplantation, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Claudia Moreno Romero
- Division of Abdominal Transplantation, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Sarah R McCarthy
- Division of Abdominal Transplantation, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Olivia L Hatton
- Department of Molecular Biology, Colorado College, Colorado Springs, CO, USA
| | - Carlos O Esquivel
- Division of Abdominal Transplantation, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Olivia M Martinez
- Division of Abdominal Transplantation, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Sheri M Krams
- Division of Abdominal Transplantation, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
29
|
Rühl J, Leung CS, Münz C. Vaccination against the Epstein-Barr virus. Cell Mol Life Sci 2020; 77:4315-4324. [PMID: 32367191 PMCID: PMC7223886 DOI: 10.1007/s00018-020-03538-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/08/2020] [Accepted: 04/21/2020] [Indexed: 12/14/2022]
Abstract
Epstein-Barr virus (EBV) was the first human tumor virus being discovered and remains to date the only human pathogen that can transform cells in vitro. 55 years of EBV research have now brought us to the brink of an EBV vaccine. For this purpose, recombinant viral vectors and their heterologous prime-boost vaccinations, EBV-derived virus-like particles and viral envelope glycoprotein formulations are explored and are discussed in this review. Even so, cell-mediated immune control by cytotoxic lymphocytes protects healthy virus carriers from EBV-associated malignancies, antibodies might be able to prevent symptomatic primary infection, the most likely EBV-associated pathology against which EBV vaccines will be initially tested. Thus, the variety of EBV vaccines reflects the sophisticated life cycle of this human tumor virus and only vaccination in humans will finally be able to reveal the efficacy of these candidates. Nevertheless, the recently renewed efforts to develop an EBV vaccine and the long history of safe adoptive T cell transfer to treat EBV-associated malignancies suggest that this oncogenic γ-herpesvirus can be targeted by immunotherapies. Such vaccination should ideally implement the very same immune control that protects healthy EBV carriers.
Collapse
Affiliation(s)
- Julia Rühl
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Winterthurerstrasse 190, CH-8057, Zürich, Switzerland
| | - Carol S Leung
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Winterthurerstrasse 190, CH-8057, Zürich, Switzerland.
| |
Collapse
|
30
|
Kaminski H, Marsères G, Cosentino A, Guerville F, Pitard V, Fournié JJ, Merville P, Déchanet-Merville J, Couzi L. Understanding human γδ T cell biology toward a better management of cytomegalovirus infection. Immunol Rev 2020; 298:264-288. [PMID: 33091199 DOI: 10.1111/imr.12922] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/04/2020] [Accepted: 09/04/2020] [Indexed: 12/28/2022]
Abstract
Cytomegalovirus (CMV) infection is responsible for significant morbidity and mortality in immunocompromised patients, namely solid organ and hematopoietic cell transplant recipients, and can induce congenital infection in neonates. There is currently an unmet need for new management and treatment strategies. Establishment of an anti-CMV immune response is critical in order to control CMV infection. The two main human T cells involved in HCMV-specific response are αβ and non-Vγ9Vδ2 T cells that belong to γδ T cell compartment. CMV-induced non-Vγ9Vδ2 T cells harbor a specific clonal expansion and a phenotypic signature, and display effector functions against CMV. So far, only two main molecular mechanisms underlying CMV sensing have been identified. Non-Vγ9Vδ2 T cells can be activated either by stress-induced surface expression of the γδT cell receptor (TCR) ligand annexin A2, or by a multimolecular stress signature composed of the γδTCR ligand endothelial protein C receptor and co-stimulatory signals such as the ICAM-1-LFA-1 axis. All this basic knowledge can be harnessed to improve the clinical management of CMV infection in at-risk patients. In particular, non-Vγ9Vδ2 T cell monitoring could help better stratify the risk of infection and move forward a personalized medicine. Moreover, recent advances in cell therapy protocols open the way for a non-Vγ9Vδ2 T cell therapy in immunocompromised patients.
Collapse
Affiliation(s)
- Hannah Kaminski
- ImmunoConcEpT UMR 5164, CNRS, Bordeaux University, Bordeaux, France.,Department of Nephrology, Transplantation, Dialysis and Apheresis, Bordeaux University Hospital, Bordeaux, France
| | - Gabriel Marsères
- ImmunoConcEpT UMR 5164, CNRS, Bordeaux University, Bordeaux, France
| | - Anaïs Cosentino
- ImmunoConcEpT UMR 5164, CNRS, Bordeaux University, Bordeaux, France.,Department of Nephrology, Transplantation, Dialysis and Apheresis, Bordeaux University Hospital, Bordeaux, France
| | - Florent Guerville
- ImmunoConcEpT UMR 5164, CNRS, Bordeaux University, Bordeaux, France.,CHU Bordeaux, Pôle de gérontologie, Bordeaux, Bordeaux, France
| | - Vincent Pitard
- ImmunoConcEpT UMR 5164, CNRS, Bordeaux University, Bordeaux, France
| | - Jean-Jacques Fournié
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR1037 INSERM, Université Toulouse III: Paul-Sabatier, ERL5294 CNRS, Université de Toulouse, Toulouse, France
| | - Pierre Merville
- ImmunoConcEpT UMR 5164, CNRS, Bordeaux University, Bordeaux, France.,Department of Nephrology, Transplantation, Dialysis and Apheresis, Bordeaux University Hospital, Bordeaux, France
| | | | - Lionel Couzi
- ImmunoConcEpT UMR 5164, CNRS, Bordeaux University, Bordeaux, France.,Department of Nephrology, Transplantation, Dialysis and Apheresis, Bordeaux University Hospital, Bordeaux, France
| |
Collapse
|
31
|
Münz C. Probing Reconstituted Human Immune Systems in Mice With Oncogenic γ-Herpesvirus Infections. Front Immunol 2020; 11:581419. [PMID: 33013936 PMCID: PMC7509489 DOI: 10.3389/fimmu.2020.581419] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 08/17/2020] [Indexed: 12/22/2022] Open
Abstract
Mice with reconstituted human immune systems can mount cell-mediated immune responses against the human tumor viruses Epstein Barr virus (EBV) and Kaposi sarcoma associated herpesvirus (KSHV). Primarily cytotoxic lymphocytes protect the vast majority of persistently infected carriers of these tumor viruses from the respective malignancies for life. Thus, EBV and KSHV infection can teach us how this potent immune control is induced, what phenotype and functions characterize the protective lymphocyte compartments and if similar immune responses could be induced by vaccination. This review will summarize similarities and differences between EBV and KSHV associated pathologies and their immune control in patients and mice with reconstituted human immune systems. Furthermore, it will high-light which aspects of the near perfect immune control can be modeled in the latter preclinical animal models and discuss their relevance for cancer immunology in general.
Collapse
Affiliation(s)
- Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, Zurich, Switzerland
| |
Collapse
|
32
|
Cytotoxicity in Epstein Barr virus specific immune control. Curr Opin Virol 2020; 46:1-8. [PMID: 32771660 DOI: 10.1016/j.coviro.2020.07.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/09/2020] [Accepted: 07/14/2020] [Indexed: 02/08/2023]
Abstract
Epstein Barr virus (EBV) is the most common human tumor virus, persistently infecting more than 95% of the human adult population and readily transforming human B cell in culture. Fortunately, only a small minority of EBV carriers develops virus associated malignancies. The majority controls persistent EBV infection with cytotoxic lymphocytes, mainly NK, γδ and CD8+ T cells and the characteristics of the required immune responses get more and more defined by primary immunodeficiencies that affect molecules of these cytotoxic lymphocytes and their investigation in mice with reconstituted human immune system components (humanized mice) that are susceptible to EBV infection and associated lymphomagenesis. The gained information should be able to guide us to develop immunotherapies against EBV and tumors in general.
Collapse
|
33
|
Biomarkers for PTLD diagnosis and therapies. Pediatr Nephrol 2020; 35:1173-1181. [PMID: 31240394 DOI: 10.1007/s00467-019-04284-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/14/2019] [Accepted: 05/31/2019] [Indexed: 02/08/2023]
Abstract
Post-transplant lymphoproliferative disorder (PTLD) represents a spectrum of lymphoproliferative disorders and is a serious complication of pediatric transplantation. The majority of PTLD are associated with Epstein Barr virus (EBV) and the characteristic EBV+ B cell lymphomas are the leading post-transplant malignancy in children. EBV+ PTLD remains a formidable issue in pediatric transplantation and is thought to result from impaired immunity to EBV as a result of immunosuppression. However, the key viral and immune factors that determine whether EBV+ PTLD develops remain unknown. Recently, there has been much interest in developing biomarkers in order to improve and achieve more personalized approaches, in the clinical diagnosis, management, and treatment of EBV+ PTLD. Here, we review the status of immune-, viral-, and B cell lymphoma-derived candidates for biomarkers of EBV+ PTLD.
Collapse
|
34
|
Djaoud Z, Parham P. Dimorphism in the TCRγ-chain repertoire defines 2 types of human immunity to Epstein-Barr virus. Blood Adv 2020; 4:1198-1205. [PMID: 32211881 PMCID: PMC7160271 DOI: 10.1182/bloodadvances.2019001179] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 02/22/2020] [Indexed: 02/02/2023] Open
Abstract
Humans form 2 groups based on their innate immunity to Epstein-Barr virus (EBV). Group 1 makes a strong natural killer (NK)-cell and γδ T-cell response, whereas group 2 makes a strong NK-cell response, but a weak γδ T-cell response. To investigate the underlying basis for this difference in γδ T-cell immunity to EBV, we used next-generation sequencing to compare the γδ T-cell receptor (TCR) repertoires of groups 1 and 2. In the absence of EBV, group 1 TCRγ chains are enriched for complementarity determining region 3 (CDR3s) containing JγP, whereas group 2 TCRγ chains are enriched for CDR3s containing Jγ2. In group 1 donors, EBV activates many γδ T cells expressing Vγ9JγP, inducing proliferation that produces a large population of activated effector cells. The TCRs using Vγ9JγP are closely related to the TCRs of γδ T cells that respond to phosphoantigens. In group 2 donors, EBV activates a small subpopulation of γδ T cells, most expressing Vγ9JγP. In conclusion, we find that differences in the TCRγ-chain repertoire underlie the differential response of group 1 and group 2 to EBV.
Collapse
Affiliation(s)
- Zakia Djaoud
- Department of Structural Biology and
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA
| | - Peter Parham
- Department of Structural Biology and
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
35
|
McHugh D, Caduff N, Murer A, Engelmann C, Deng Y, Zdimerova H, Zens K, Chijioke O, Münz C. Infection and immune control of human oncogenic γ-herpesviruses in humanized mice. Philos Trans R Soc Lond B Biol Sci 2020; 374:20180296. [PMID: 30955487 DOI: 10.1098/rstb.2018.0296] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Epstein-Barr virus (EBV) and Kaposi sarcoma-associated herpesvirus (KSHV) comprise the oncogenic human γ-herpesvirus family and are responsible for 2-3% of all tumours in man. With their prominent growth-transforming abilities and high prevalence in the human population, these pathogens have probably shaped the human immune system throughout evolution for near perfect immune control of the respective chronic infections in the vast majority of healthy pathogen carriers. The exclusive tropism of EBV and KSHV for humans has, however, made it difficult in the past to study their infection, tumourigenesis and immune control in vivo. Mice with reconstituted human immune system components (humanized mice) support replication of both viruses with both persisting latent and productive lytic infection. Moreover, B-cell lymphomas can be induced by EBV alone and KSHV co-infection with gene expression hallmarks of human malignancies that are associated with both viruses. Furthermore, cell-mediated immune control by primarily cytotoxic lymphocytes is induced upon infection and can be probed for its functional characteristics as well as putative requirements for its priming. Insights that have been gained from this model and remaining questions will be discussed in this review. This article is part of the theme issue 'Silent cancer agents: multi-disciplinary modelling of human DNA oncoviruses'.
Collapse
Affiliation(s)
- Donal McHugh
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| | - Nicole Caduff
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| | - Anita Murer
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| | - Christine Engelmann
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| | - Yun Deng
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| | - Hana Zdimerova
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| | - Kyra Zens
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| | - Obinna Chijioke
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich , Switzerland
| |
Collapse
|
36
|
Latour S, Fischer A. Signaling pathways involved in the T-cell-mediated immunity against Epstein-Barr virus: Lessons from genetic diseases. Immunol Rev 2020; 291:174-189. [PMID: 31402499 DOI: 10.1111/imr.12791] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/05/2019] [Accepted: 06/10/2019] [Indexed: 12/13/2022]
Abstract
Primary immunodeficiencies (PIDs) provide researchers with unique models to understand in vivo immune responses in general and immunity to infections in particular. In humans, impaired immune control of Epstein-Barr virus (EBV) infection is associated with the occurrence of several different immunopathologic conditions; these include non-malignant and malignant B-cell lymphoproliferative disorders, hemophagocytic lymphohistiocytosis (HLH), a severe inflammatory condition, and a chronic acute EBV infection of T cells. Studies of PIDs associated with a predisposition to develop severe, chronic EBV infections have led to the identification of key components of immunity to EBV - notably the central role of T-cell expansion and its regulation in the pathophysiology of EBV-associated diseases. On one hand, the defective expansion of EBV-specific CD8 T cells results from mutations in genes involved in T-cell activation (such as RASGRP1, MAGT1, and ITK), DNA metabolism (CTPS1) or co-stimulatory pathways (CD70, CD27, and TNFSFR9 (also known as CD137/4-1BB)) leads to impaired elimination of proliferating EBV-infected B cells and the occurrence of lymphoma. On the other hand, protracted T-cell expansion and activation after the defective killing of EBV-infected B cells is caused by genetic defects in the components of the lytic granule exocytosis pathway or in the small adapter protein SH2D1A (also known as SAP), a key activator of T- and NK cell-cytotoxicity. In this setting, the persistence of EBV-infected cells results in HLH, a condition characterized by unleashed T-cell and macrophage activation. Moreover, genetic defects causing selective vulnerability to EBV infection have highlighted the role of co-receptor molecules (CD27, CD137, and SLAM-R) selectively involved in immune responses against infected B cells via specific T-B cell interactions.
Collapse
Affiliation(s)
- Sylvain Latour
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, Inserm UMR 1163, Paris, France.,University Paris Descartes Sorbonne Paris Cité, Imagine Institut, Paris, France
| | - Alain Fischer
- University Paris Descartes Sorbonne Paris Cité, Imagine Institut, Paris, France.,Department of Pediatric Immunology, Hematology and Rheumatology, Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Paris, France.,Collège de France, Paris, France.,Inserm UMR 1163, Paris, France
| |
Collapse
|
37
|
M A, Chatterjee S, A P, S M, Davuluri S, Ar AK, T A, M P, Cs P, Sinha M, Chugani A, R VP, Kk A, R S J. Natural Killer cell transcriptome during primary EBV infection and EBV associated Hodgkin Lymphoma in children-A preliminary observation. Immunobiology 2020; 225:151907. [PMID: 32044149 DOI: 10.1016/j.imbio.2020.151907] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/09/2020] [Accepted: 01/25/2020] [Indexed: 01/22/2023]
Abstract
Epstein Barr Viral infection is a common childhood infection in India and is also nearly 100 % etiologically associated with pediatric Hodgkin Lymphoma (HL). The main question in EBV immunobiology has been, why only a small subset of infected individuals develop EBV associated malignancies, while the vast majority carry this virus asymptomatically for life. Natural Killer (NK) cells, with a phenotype of CD56dim CD16+ exhibit potent cytotoxicity towards both virus infected cells and transformed cells and hence have been considered to be crucial in preventing the development of symptomatic EBV infection and lymphoma. In order to get an insight into the various possible molecular aspects of NK cells, in the pathogenesis of both these EBV mediated diseases in children we studied the whole transcriptome of MACS sorted CD56dim CD16 + NK cells from four patients from each of the three groups of children viz. Infectious Mononucleosis (IM), HL and age matched controls by using a massively parallel sequencing approach. NK cells from both IM and HL had down-regulated innate immunity and chemokine signaling genes. While down-regulation of genes responsible for polarization of the secretory apparatus, activated NK cell signaling and MAP kinase signaling were exclusive to NK cells in patients with IM, in NK cells of HL, specifically, genes involved in extracellular matrix (ECM) - receptor interaction, cytokine-cytokine receptor interaction, TNF signaling, Toll-like receptor signaling pathway and cytosolic DNA-sensing pathways were significantly down-regulated. Enrichment analysis showed STAT3 to be the most significant transcription factor (TF) for the down-regulated genes in IM, whereas, GATA1 was found to be the most significant TF for the genes down-regulated in HL. Analysis of protein interaction network identified functionally important protein clusters. Top clusters, comprised of down-regulated genes, involved in signaling and ubiquitin-related processes and pathways. These may perhaps be responsible for the hypo-responsiveness of NK cells in both diseases. These possibly point to different deficiencies in NK cell activation, loss of activating receptor signaling and degranulation in IM, versus loss of cytokine and chemokine signaling in HL, in the two EBV associated pathologies investigated. Various suppressed molecules and pathways were novel, which have not been reported earlier and could therefore be potential targets for immunotherapy of NK cell reactivation in both the diseases in future.
Collapse
Affiliation(s)
- Alka M
- Departments of Microbiology, Kidwai Memorial Institute of Oncology, Bangalore, India
| | | | - Parchure A
- Departments of Microbiology, Kidwai Memorial Institute of Oncology, Bangalore, India
| | - Mahantesh S
- Departments of Microbiology, Indira Gandhi Institute of Child Health, Bangalore, India
| | - Sravanthi Davuluri
- Biological Data Analyzers' Association (BdataA), Electronic City, Phase I, Bangalore, India
| | - Arun Kumar Ar
- Departments of Pediatric Oncology, Kidwai Memorial Institute of Oncology, India
| | - Avinash T
- Departments of Pediatric Oncology, Kidwai Memorial Institute of Oncology, India
| | - Padma M
- Departments of Pediatric Oncology, Kidwai Memorial Institute of Oncology, India
| | - Premalata Cs
- Departments of Pathology, Kidwai Memorial Institute of Oncology, Bangalore, India
| | - Mahua Sinha
- Departments of Microbiology, Kidwai Memorial Institute of Oncology, Bangalore, India
| | | | | | - Acharya Kk
- Biological Data Analyzers' Association (BdataA), Electronic City, Phase I, Bangalore, India; Institute of Bioinformatics and Applied Biotechnology, Bangalore, India
| | - Jayshree R S
- Departments of Microbiology, Kidwai Memorial Institute of Oncology, Bangalore, India.
| |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW Natural killer (NK) cells are effector cells of the innate immune system that can lyse target cells without prior sensitization and are important in host defense to virally infected and transformed cells. Although the concept of 'missing-self' would suggest NK cells could target foreign allografts, the prevailing dogma has been that NK cells are not active participants in the rejection of solid organ allografts. This review summarizes recent studies that challenge this conclusion and instead suggest NK cells are important in outcomes posttransplant. RECENT FINDINGS NK cells expressing specific cell surface receptors may promote graft damage and rejection. However, recent studies suggest some NK cell subsets have tolerogenic or immunoregulatory potential and promote graft stability, suggesting a dichotomous role for NK cells after transplant. Furthermore, NK cells respond to cells infected with cytomegalovirus and Epstein-Barr virus, and studies suggest some NK cells have immune memory. SUMMARY Our understanding of the role of NK cells posttransplant has evolved from 'no role' to the current idea that NK cells may have 'complex interactions' that impact graft outcomes. Additional studies, using cutting edge techniques to comprehensively analyze the phenotypic and functional subsets of NK cells in transplant recipients, are clearly necessary.
Collapse
|
39
|
Sant S, Jenkins MR, Dash P, Watson KA, Wang Z, Pizzolla A, Koutsakos M, Nguyen TH, Lappas M, Crowe J, Loudovaris T, Mannering SI, Westall GP, Kotsimbos TC, Cheng AC, Wakim L, Doherty PC, Thomas PG, Loh L, Kedzierska K. Human γδ T-cell receptor repertoire is shaped by influenza viruses, age and tissue compartmentalisation. Clin Transl Immunology 2019; 8:e1079. [PMID: 31559018 PMCID: PMC6756999 DOI: 10.1002/cti2.1079] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/16/2019] [Accepted: 08/17/2019] [Indexed: 12/21/2022] Open
Abstract
Background Although γδ T cells comprise up to 10% of human peripheral blood T cells, questions remain regarding their role in disease states and T‐cell receptor (TCR) clonal expansions. We dissected anti‐viral functions of human γδ T cells towards influenza viruses and defined influenza‐reactive γδ TCRs in the context of γδ‐TCRs across the human lifespan. Methods We performed 51Cr‐killing assay and single‐cell time‐lapse live video microscopy to define mechanisms underlying γδ T‐cell‐mediated killing of influenza‐infected targets. We assessed cytotoxic profiles of γδ T cells in influenza‐infected patients and IFN‐γ production towards influenza‐infected lung epithelial cells. Using single‐cell RT‐PCR, we characterised paired TCRγδ clonotypes for influenza‐reactive γδ T cells in comparison with TCRs from healthy neonates, adults, elderly donors and tissues. Results We provide the first visual evidence of γδ T‐cell‐mediated killing of influenza‐infected targets and show distinct features to those reported for CD8+ T cells. γδ T cells displayed poly‐cytotoxic profiles in influenza‐infected patients and produced IFN‐γ towards influenza‐infected cells. These IFN‐γ‐producing γδ T cells were skewed towards the γ9δ2 TCRs, particularly expressing the public GV9‐TCRγ, capable of pairing with numerous TCR‐δ chains, suggesting their significant role in γδ T‐cell immunity. Neonatal γδ T cells displayed extensive non‐overlapping TCRγδ repertoires, while adults had enriched γ9δ2‐pairings with diverse CDR3γδ regions. Conversely, the elderly showed distinct γδ‐pairings characterised by large clonal expansions, a profile also prominent in adult tissues. Conclusion Human TCRγδ repertoire is shaped by age, tissue compartmentalisation and the individual's history of infection, suggesting that these somewhat enigmatic γδ T cells indeed respond to antigen challenge.
Collapse
Affiliation(s)
- Sneha Sant
- Department of Microbiology and Immunology University of Melbourne at The Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia
| | - Misty R Jenkins
- Immunology Division Walter and Eliza Hall Institute Melbourne VIC Australia.,LaTrobe Institute for Molecular Science La Trobe University Melbourne VIC Australia.,Department of Medical Biology The University of Melbourne Melbourne VIC Australia
| | - Pradyot Dash
- Department of Immunology St Jude Children's Research Hospital Memphis TN USA
| | - Katherine A Watson
- Immunology Division Walter and Eliza Hall Institute Melbourne VIC Australia
| | - Zhongfang Wang
- Department of Microbiology and Immunology University of Melbourne at The Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia
| | - Angela Pizzolla
- Department of Microbiology and Immunology University of Melbourne at The Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia
| | - Marios Koutsakos
- Department of Microbiology and Immunology University of Melbourne at The Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia
| | - Thi Ho Nguyen
- Department of Microbiology and Immunology University of Melbourne at The Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia
| | - Martha Lappas
- Obstetrics, Nutrition and Endocrinology Group Department of Obstetrics & Gynaecology Mercy Hospital for Women University of Melbourne Melbourne VIC Australia
| | | | - Tom Loudovaris
- Immunology and Diabetes Unit St Vincent's Institute of Medical Research Fitzroy VIC Australia
| | - Stuart I Mannering
- Immunology and Diabetes Unit St Vincent's Institute of Medical Research Fitzroy VIC Australia
| | - Glen P Westall
- Lung Transplant Unit Alfred Hospital Melbourne VIC Australia
| | - Tom C Kotsimbos
- Department of Allergy, Immunology and Respiratory Medicine The Alfred Hospital Melbourne VIC Australia.,Department of Medicine Central Clinical School The Alfred Hospital Melbourne Monash University Melbourne VIC Australia
| | - Allen C Cheng
- School of Public Health and Preventive Medicine Monash University Melbourne VIC Australia.,Infection Prevention and Healthcare Epidemiology Unit Alfred Health Melbourne VIC Australia
| | - Linda Wakim
- Department of Microbiology and Immunology University of Melbourne at The Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia
| | - Peter C Doherty
- Department of Microbiology and Immunology University of Melbourne at The Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia.,Immunology Division Walter and Eliza Hall Institute Melbourne VIC Australia
| | - Paul G Thomas
- Department of Immunology St Jude Children's Research Hospital Memphis TN USA
| | - Liyen Loh
- Department of Microbiology and Immunology University of Melbourne at The Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology University of Melbourne at The Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia
| |
Collapse
|
40
|
Long HM, Meckiff BJ, Taylor GS. The T-cell Response to Epstein-Barr Virus-New Tricks From an Old Dog. Front Immunol 2019; 10:2193. [PMID: 31620125 PMCID: PMC6759930 DOI: 10.3389/fimmu.2019.02193] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 08/30/2019] [Indexed: 11/13/2022] Open
Abstract
Epstein-Barr virus (EBV) infects most people and establishes life-long infection controlled by the host's immune system. The genetic stability of the virus, deep understanding of the viral antigens and immune epitopes recognized by the host's T-cell system and the fact that recent infection can be identified by the development of symptomatic infectious mononucleosis makes EBV a powerful system in which to study human immunology. The association between EBV and multiple cancers also means that the lessons learned have strong translational potential. Increasing evidence of a role for resident memory T-cells and non-conventional γδ T-cells in controlling EBV infection suggests new opportunities for research and means the virus will continue to provide exciting new insights into human biology and immunology into the future.
Collapse
Affiliation(s)
- Heather M. Long
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | | | - Graham S. Taylor
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
41
|
Epstein-Barr virus-related post-transplant lymphoproliferative disease (EBV-PTLD) in the setting of allogeneic stem cell transplantation: a comprehensive review from pathogenesis to forthcoming treatment modalities. Bone Marrow Transplant 2019; 55:25-39. [DOI: 10.1038/s41409-019-0548-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/10/2019] [Accepted: 04/15/2019] [Indexed: 12/17/2022]
|
42
|
Damania B, Münz C. Immunodeficiencies that predispose to pathologies by human oncogenic γ-herpesviruses. FEMS Microbiol Rev 2019; 43:181-192. [PMID: 30649299 PMCID: PMC6435449 DOI: 10.1093/femsre/fuy044] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/30/2018] [Indexed: 12/13/2022] Open
Abstract
Human γ-herpesviruses include the closely related tumor viruses Epstein Barr virus (EBV) and Kaposi sarcoma-associated herpesvirus (KSHV). EBV is the most growth-transforming pathogen known and is linked to at least seven human malignancies. KSHV is also associated with three human cancers. Most EBV- and KSHV-infected individuals fortunately remain disease-free despite persistent infection and this is likely due to the robustness of the immune control that they mount against these tumor viruses. However, upon immune suppression EBV- and KSHV-associated malignancies emerge at increased frequencies. Moreover, primary immunodeficiencies with individual mutations that predispose to EBV or KSHV disease allow us to gain insights into a catalog of molecules that are required for the immune control of these tumor viruses. Curiously, there is little overlap between the mutation targets that predispose individuals to EBV versus KSHV disease, even so both viruses can infect the same host cell, human B cells. These differences will be discussed in this review. A better understanding of the crucial components in the near-perfect life-long immune control of EBV and KSHV should allow us to target malignancies that are associated with these viruses, but also induce similar immune responses against other tumors.
Collapse
Affiliation(s)
- Blossom Damania
- Lineberger Cancer Research Center and Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, 8057 Zürich, Switzerland
| |
Collapse
|
43
|
Jia ZH, Li YY, Wang JY, Zhang JY, Huang A, Guo XD, Zhu ZY, Wang FS, Wu XL. Activated γδ T cells exhibit cytotoxicity and the capacity for viral clearance in patients with acute hepatitis B. Clin Immunol 2019; 202:40-48. [PMID: 30914281 DOI: 10.1016/j.clim.2019.03.005] [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: 09/11/2018] [Revised: 01/16/2019] [Accepted: 03/22/2019] [Indexed: 12/21/2022]
Abstract
γδ T cells are a unique population of lymphocytes that have regulatory roles in patients with chronic hepatitis B (CHB); however, their role in acute hepatitis B (AHB) infection remains unclear. Phenotype and function of γδ T cells were analyzed in 29 AHB patients, 28 CHB patients, and 30 healthy controls (HCs) using immunofunctional assays. Compared with HCs and CHB patients, decreased peripheral and increased hepatic γδ T cells were found in AHB patients. Increased hepatic γδ T cells in AHB patients were attributed to elevated hepatic chemokine levels. Peripheral γδ T cells exhibited highly activated and terminally differentiated memory phenotype in AHB patients. Consistently, peripheral γδ T cells in AHB patients showed increased cytotoxic capacity and enhanced antiviral activity which was further proved in longitudinal study. Activated γδ T cells in AHB patients exhibited increased cytotoxicity and capacity for viral clearance associated with liver injury and the control of infection.
Collapse
Affiliation(s)
- Zheng-Hu Jia
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yuan-Yuan Li
- Treatment and Research Center for Infectious Diseases, Beijing 302 Hospital, Beijing 100039, China
| | - Jing-Ya Wang
- Department of Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Heping, Tianjin, China
| | - Ji-Yuan Zhang
- Treatment and Research Center for Infectious Diseases, Beijing 302 Hospital, Beijing 100039, China
| | - Ang Huang
- The Center for Non-infectious Liver Diseases, Beijing 302 Hospital, 100039, China
| | - Xiao-Dong Guo
- Department of Pathology, Beijing 302 Hospital, 100039, China
| | - Zhen-Yu Zhu
- Hepatobiliary Surgery Center, Beijing 302 Hospital, 100039, China
| | - Fu-Sheng Wang
- Treatment and Research Center for Infectious Diseases, Beijing 302 Hospital, Beijing 100039, China.
| | - Xiao-Li Wu
- School of Life Sciences, Tianjin University, Tianjin Engineering Center of Micro Nano Biomaterials and Detection Treatment Technology, Collaborative Innovation Center of Chemical Science and Engineering, Tianjin 300072, China.
| |
Collapse
|
44
|
Hildreth AD, O'Sullivan TE. Tissue-Resident Innate and Innate-Like Lymphocyte Responses to Viral Infection. Viruses 2019; 11:v11030272. [PMID: 30893756 PMCID: PMC6466361 DOI: 10.3390/v11030272] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/13/2019] [Accepted: 03/14/2019] [Indexed: 12/16/2022] Open
Abstract
Infection is restrained by the concerted activation of tissue-resident and circulating immune cells. Recent discoveries have demonstrated that tissue-resident lymphocyte subsets, comprised of innate lymphoid cells (ILCs) and unconventional T cells, have vital roles in the initiation of primary antiviral responses. Via direct and indirect mechanisms, ILCs and unconventional T cell subsets play a critical role in the ability of the immune system to mount an effective antiviral response through potent early cytokine production. In this review, we will summarize the current knowledge of tissue-resident lymphocytes during initial viral infection and evaluate their redundant or nonredundant contributions to host protection or virus-induced pathology.
Collapse
Affiliation(s)
- Andrew D Hildreth
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 900953, USA.
| | - Timothy E O'Sullivan
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 900953, USA.
| |
Collapse
|
45
|
Abstract
Epstein–Barr virus (EBV) contributes to about 1.5% of all cases of human cancer worldwide, and viral genes are expressed in the malignant cells. EBV also very efficiently causes the proliferation of infected human B lymphocytes. The functions of the viral proteins and small RNAs that may contribute to EBV-associated cancers are becoming increasingly clear, and a broader understanding of the sequence variation of the virus genome has helped to interpret their roles. The improved understanding of the mechanisms of these cancers means that there are great opportunities for the early diagnosis of treatable stages of EBV-associated cancers and the use of immunotherapy to target EBV-infected cells or overcome immune evasion. There is also scope for preventing disease by immunization and for developing therapeutic agents that target the EBV gene products expressed in the cancers.
Collapse
Affiliation(s)
- Paul J. Farrell
- Section of Virology, Imperial College Faculty of Medicine, London W2 1PG, United Kingdom
| |
Collapse
|
46
|
Zhao Y, Lin L, Xiao Z, Li M, Wu X, Li W, Li X, Zhao Q, Wu Y, Zhang H, Yin J, Zhang L, Cho CH, Shen J. Protective Role of γδ T Cells in Different Pathogen Infections and Its Potential Clinical Application. J Immunol Res 2018; 2018:5081634. [PMID: 30116753 PMCID: PMC6079409 DOI: 10.1155/2018/5081634] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/10/2018] [Indexed: 12/24/2022] Open
Abstract
γδ T cells, a subgroup of T cells based on the γδ TCR, when compared with conventional T cells (αβ T cells), make up a very small proportion of T cells. However, its various subgroups are widely distributed in different parts of the human body and are attractive effectors for infectious disease immunity. γδ T cells are activated and expanded by nonpeptidic antigens (P-Ags), major histocompatibility complex (MHC) molecules, and lipids which are associated with different kinds of pathogen infections. Activation and proliferation of γδ T cells play a significant role in diverse infectious diseases induced by viruses, bacteria, and parasites and exert their potential effector function to effectively eliminate infection. It is well known that many types of infectious diseases are detrimental to human life and health and give rise to high incidence of illnesses and death rate all over the world. To date, there is no comprehensive understanding of the correlation between γδ T cells and infectious diseases. In this review, we will focus on the various subgroups of γδ T cells (mainly Vδ1 T cells and Vδ2 T cells) which can induce multiple immune responses or effective functions to fight against common pathogen infections, such as Mycobacterium tuberculosis, Listeria monocytogenes, influenza viruses, HIV, EBV, and HBV. Hopefully, the gamma-delta T cell study will provide a novel effective way to treat infectious diseases.
Collapse
Affiliation(s)
- Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Ling Lin
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Wanping Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaobing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Qijie Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yuanlin Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Hanyu Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Jianhua Yin
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Lingling Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Chi Hin Cho
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
47
|
Latour S, Winter S. Inherited Immunodeficiencies With High Predisposition to Epstein-Barr Virus-Driven Lymphoproliferative Diseases. Front Immunol 2018; 9:1103. [PMID: 29942301 PMCID: PMC6004768 DOI: 10.3389/fimmu.2018.01103] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 05/02/2018] [Indexed: 01/16/2023] Open
Abstract
Epstein–Barr Virus (EBV) is a gamma-herpes virus that infects 90% of humans without any symptoms in most cases, but has an oncogenic potential, especially in immunocompromised individuals. In the past 30 years, several primary immunodeficiencies (PIDs) associated with a high risk to develop EBV-associated lymphoproliferative disorders (LPDs), essentially consisting of virus-associated hemophagocytic syndrome, non-malignant and malignant B-cell LPDs including non-Hodgkin and Hodgkin’s types of B lymphomas have been characterized. Among them are SH2D1A (SAP), XIAP, ITK, MAGT1, CD27, CD70, CTPS1, RASGRP1, and CORO1A deficiencies. Penetrance of EBV infection ranges from 50 to 100% in those PIDs. Description of large cohorts and case reports has refined the specific phenotypes associated with these PIDs helping to the diagnosis. Specific pathways required for protective immunity to EBV have emerged from studies of these PIDs. SLAM-associated protein-dependent SLAM receptors and MAGT1-dependent NKG2D pathways are important for T and NK-cell cytotoxicity toward EBV-infected B-cells, while CD27–CD70 interactions are critical to drive the expansion of EBV-specific T-cells. CTPS1 and RASGRP1 deficiencies further strengthen that T-lymphocyte expansion is a key step in the immune response to EBV. These pathways appear to be also important for the anti-tumoral immune surveillance of abnormal B cells. Monogenic PIDs should be thus considered in case of any EBV-associated LPDs.
Collapse
Affiliation(s)
- Sylvain Latour
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Paris, France.,Imagine Institute, Paris Descartes University, Sorbonne Paris Cité, Paris, France.,Equipe de Recherche Labéllisée, Ligue National contre le Cancer, Paris, France
| | - Sarah Winter
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Paris, France.,Imagine Institute, Paris Descartes University, Sorbonne Paris Cité, Paris, France.,Equipe de Recherche Labéllisée, Ligue National contre le Cancer, Paris, France
| |
Collapse
|
48
|
de Witte MA, Sarhan D, Davis Z, Felices M, Vallera DA, Hinderlie P, Curtsinger J, Cooley S, Wagner J, Kuball J, Miller JS. Early Reconstitution of NK and γδ T Cells and Its Implication for the Design of Post-Transplant Immunotherapy. Biol Blood Marrow Transplant 2018; 24:1152-1162. [PMID: 29505821 PMCID: PMC5993609 DOI: 10.1016/j.bbmt.2018.02.023] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/26/2018] [Indexed: 12/18/2022]
Abstract
Relapse is the most frequent cause of treatment failure after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Natural killer (NK) cells and γδ T cells reconstitute early after allo-HSCT, contribute to tumor immunosurveillance via major histocompatibility complex-independent mechanisms and do not induce graft-versus-host disease. Here we performed a quantitative and qualitative analysis of the NK and γδ T cell repertoire in healthy individuals, recipients of HLA-matched sibling or unrelated donor allo-HSCT (MSD/MUD-HSCT) and umbilical cord blood-HSCT (UCB-HSCT). NK cells are present at high frequencies in all allo-HSCT recipients. Immune reconstitution (IR) of vδ2+ cells depended on stem cell source. In MSD/MUD-HSCT recipients, vδ2+ comprise up to 8% of the total lymphocyte pool, whereas vδ2+ T cells are barely detectable in UCB-HSCT recipients. Vδ1+ IR was driven by CMV reactivation and was comparable between MSD/MUD-HSCT and UCB-HSCT. Strategies to augment NK cell mediated tumor responses, similar to IL-15 and antibodies, also induced vδ2+ T cell responses against a variety of different tumor targets. Vδ1+ γδ T cells were induced less by these same stimuli. We also identified elevated expression of the checkpoint inhibitory molecule TIGIT (T cell Ig and ITIM domain), which is also observed on tumor-infiltrating lymphocytes and epidermal γδ T cells. Collectively, these data show multiple strategies that can result in a synergized NK and γδ T cell antitumor response. In the light of recent developments of low-toxicity allo-HSCT platforms, these interventions may contribute to the prevention of early relapse.
Collapse
Affiliation(s)
- Moniek A de Witte
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota; Department of Hematology, Cancer Center, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Dhifaf Sarhan
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Zachary Davis
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Martin Felices
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Daniel A Vallera
- Department of Therapeutic Radiology-Radiation Oncology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Peter Hinderlie
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Julie Curtsinger
- Translational Therapy Laboratory, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Sarah Cooley
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - John Wagner
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - Jurgen Kuball
- Department of Hematology, Cancer Center, University Medical Centre Utrecht, Utrecht, the Netherlands; Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Jeffrey S Miller
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
49
|
Wilk AJ, Blish CA. Diversification of human NK cells: Lessons from deep profiling. J Leukoc Biol 2018; 103:629-641. [PMID: 29350874 PMCID: PMC6133712 DOI: 10.1002/jlb.6ri0917-390r] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/06/2017] [Accepted: 12/29/2017] [Indexed: 12/14/2022] Open
Abstract
NK cells are innate lymphocytes with important roles in immunoregulation, immunosurveillance, and cytokine production. Originally defined on the functional basis of their "natural" ability to lyse tumor targets and thought to be a relatively homogeneous group of lymphocytes, NK cells possess a remarkable degree of phenotypic and functional diversity due to the combinatorial expression of an array of activating and inhibitory receptors. Diversification of NK cells is multifaceted: mechanisms of NK cell education that promote self-tolerance result in a heterogeneous repertoire that further diversifies upon encounters with viral pathogens. Here, we review the genetic, developmental, and environmental sources of NK cell diversity with a particular focus on deep profiling and single-cell technologies that will enable a more thorough and accurate dissection of this intricate and poorly understood lymphocyte lineage.
Collapse
Affiliation(s)
- Aaron J. Wilk
- Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Catherine A. Blish
- Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, and Stanford Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| |
Collapse
|
50
|
Abstract
Epstein-Barr virus (EBV) infects about 90% of adults worldwide. It is the main cause of infectious mononucleosis, which is observed most frequently in adolescents. The disease can last several weeks and is characterized by lymphocytosis, sore throat, lymphadenopathy, and fatigue. Exposure to oral secretions during deep kissing has been identified as the major source for primary EBV infection in adolescents. Oral secretions are also thought to be the source for younger children through intimate intact or sharing food and eating utensils, although this has not been confirmed. Unlike most acute viral illnesses such as influenza, the incubation period of symptomatic primary EBV infection is unusually long, lasting about six weeks. Diagnosis is typically made by heterophile antibody tests and/or EBV-specific antibody tests. Long-term consequences may result from acquisition of the virus, including nasopharyngeal carcinoma and lymphomas. Nevertheless, there remains a surprising dearth of knowledge regarding the establishment of an immune response to persistent EBV infection, especially during the incubation period. This lack of knowledge has impaired our ability to develop an effective prophylactic EBV vaccine, despite various attempts. Our greatest challenges in EBV research are to develop a prophylactic vaccine and devise treatment strategies for persons already infected with EBV.
Collapse
Affiliation(s)
| | - Priya S Verghese
- Department of Pediatrics, University of Minnesota Medical Center, Minneapolis, MN 55455, USA
| | - Henry H Balfour
- Department of Laboratory Medicine and Pathology, USA; Department of Pediatrics, University of Minnesota Medical Center, Minneapolis, MN 55455, USA.
| |
Collapse
|