1
|
Sturmlechner I, Jain A, Hu B, Jadhav RR, Cao W, Okuyama H, Tian L, Weyand CM, Goronzy JJ. Aging trajectories of memory CD8 + T cells differ by their antigen specificity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.26.605197. [PMID: 39211225 PMCID: PMC11360919 DOI: 10.1101/2024.07.26.605197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Memory T cells are a highly dynamic and heterogeneous population that is maintained by cytokine-driven homeostatic proliferation interspersed with episodes of antigen-mediated expansion and contraction which affect their functional state and their durability. This heterogeneity complicates studies on the impact of aging on global human memory cells, specifically, it is unclear how aging drives memory T cell dysfunction. Here, we used chronic infection with Epstein-Barr virus (EBV) to assess the influence of age on memory states at the level of antigen-specific CD8 + T cells. We find that in young adults (<40 years), EBV-specific CD8 + T cells assume preferred differentiation states depending on their peptide specificity. By age >65-years, different T cell specificities had undergone largely distinct aging trajectories, which had in common a loss in adaptive and a gain in innate immunity signatures. No evidence was seen for cellular senescence or exhaustion. While naïve/stem-like EBV-specific T cells disappeared with age, T cell diversity of EBV-specific memory cells did not change or even increased. In summary, by controlling for antigen specificity we uncover age-associated shifts in gene expression and TCR diversity that have implications for optimizing vaccination strategies and adoptive T cell therapy.
Collapse
|
2
|
Couturaud B, Doix B, Carretero-Iglesia L, Allard M, Pradervand S, Hebeisen M, Rufer N. Overall avidity declines in TCR repertoires during latent CMV but not EBV infection. Front Immunol 2023; 14:1293090. [PMID: 38053994 PMCID: PMC10694213 DOI: 10.3389/fimmu.2023.1293090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/06/2023] [Indexed: 12/07/2023] Open
Abstract
Introduction The avidity of the T-cell receptor (TCR) for antigenic peptides presented by the MHC (pMHC) on cells is an essential parameter for efficient T cell-mediated immunity. Yet, whether the TCR-ligand avidity can drive the clonal evolution of virus antigen-specific CD8 T cells, and how this process is determined in latent Cytomegalovirus (CMV)- against Epstein-Barr virus (EBV)-mediated infection remains largely unknown. Methods To address these issues, we quantified monomeric TCR-pMHC dissociation rates on CMV- and EBV-specific individual TCRαβ clonotypes and polyclonal CD8 T cell populations in healthy donors over a follow-up time of 15-18 years. The parameters involved during the long-term persistence of virus-specific T cell clonotypes were further evaluated by gene expression profiling, phenotype and functional analyses. Results Within CMV/pp65-specific T cell repertoires, a progressive contraction of clonotypes with high TCR-pMHC avidity and low CD8 binding dependency was observed, leading to an overall avidity decline during long-term antigen exposure. We identified a unique transcriptional signature preferentially expressed by high-avidity CMV/pp65-specific T cell clonotypes, including the inhibitory receptor LILRB1. Interestingly, T cell clonotypes of high-avidity showed higher LILRB1 expression than the low-avidity ones and LILRB1 blockade moderately increased T cell proliferation. Similar findings were made for CD8 T cell repertoires specific for the CMV/IE-1 epitope. There was a gradual in vivo loss of high-avidity T cells with time for both CMV specificities, corresponding to virus-specific CD8 T cells expressing enhanced LILRB1 levels. In sharp contrast, the EBV/BMFL1-specific T cell clonal composition and distribution, once established, displayed an exceptional stability, unrelated to TCR-pMHC binding avidity or LILRB1 expression. Conclusions These findings reveal an overall long-term avidity decline of CMV- but not EBV-specific T cell clonal repertoires, highlighting the differing role played by TCR-ligand avidity over the course of these two latent herpesvirus infections. Our data further suggest that the inhibitor receptor LILRB1 potentially restricts the clonal expansion of high-avidity CMV-specific T cell clonotypes during latent infection. We propose that the mechanisms regulating the long-term outcome of CMV- and EBV-specific memory CD8 T cell clonotypes in humans are distinct.
Collapse
Affiliation(s)
- Barbara Couturaud
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Bastien Doix
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Laura Carretero-Iglesia
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Mathilde Allard
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Sylvain Pradervand
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
- Lausanne Genomic Technologies Facility (LGTF), University of Lausanne, Lausanne, Switzerland
| | - Michael Hebeisen
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Nathalie Rufer
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
3
|
Yamada M, Macedo C, Louis K, Shi T, Landsittel D, Nguyen C, Shinjoh M, Michaels MG, Feingold B, Mazariegos GV, Green M, Metes D. Distinct association between chronic Epstein-Barr virus infection and T cell compartments from pediatric heart, kidney, and liver transplant recipients. Am J Transplant 2023; 23:1145-1158. [PMID: 37187296 DOI: 10.1016/j.ajt.2023.05.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 04/23/2023] [Accepted: 05/11/2023] [Indexed: 05/17/2023]
Abstract
Chronic Epstein-Barr virus (EBV) infection after pediatric organ transplantation (Tx) accounts for significant morbidity and mortality. The risk of complications, such as posttransplant lymphoproliferative disorders, in high viral load (HVL) carriers is the highest in heart Tx recipients. However, the immunologic signatures of such a risk have been insufficiently defined. Here, we assessed the phenotypic, functional, and transcriptomic profiles of peripheral blood CD8+/CD4+ T cells, including EBV-specific T cells, in 77 pediatric heart, kidney, and liver Tx recipients and established the relationship between memory differentiation and progression toward exhaustion. Unlike kidney and liver HVL carriers, heart HVL carriers displayed distinct CD8+ T cells with (1) up-regulation of interleukin-21R, (2) decreased naive phenotype and altered memory differentiation, (3) accumulation of terminally exhausted (TEX PD-1+T-bet-Eomes+) and decrease of functional precursors of exhausted (TPEX PD-1intT-bet+) effector subsets, and (4) transcriptomic signatures supporting the phenotypic changes. In addition, CD4+ T cells from heart HVL carriers displayed similar changes in naive and memory subsets, elevated Th1 follicular helper cells, and plasma interleukin-21, suggesting an alternative inflammatory mechanism that governs T cell responses in heart Tx recipients. These results may explain the different incidences of EBV complications and may help improve the risk stratification and clinical management of different types of Tx recipients.
Collapse
Affiliation(s)
- Masaki Yamada
- Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, USA; Thomas E. Starzl Transplant Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Camila Macedo
- Thomas E. Starzl Transplant Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kevin Louis
- Thomas E. Starzl Transplant Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Kidney Transplant Department, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris, Paris, France
| | - Tiange Shi
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Douglas Landsittel
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University, Indiana, Pennsylvania, USA
| | - Christina Nguyen
- Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, USA
| | - Masayoshi Shinjoh
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Marian G Michaels
- Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, USA; Thomas E. Starzl Transplant Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Brian Feingold
- Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, USA; Clinical and Translational Science, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - George V Mazariegos
- Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, USA; Thomas E. Starzl Transplant Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michael Green
- Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, USA; Thomas E. Starzl Transplant Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Diana Metes
- Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, USA; Thomas E. Starzl Transplant Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
4
|
Langan D, Wang R, Tidwell K, Mitiku S, Farrell A, Johnson C, Parks A, Suarez L, Jain S, Kim S, Jones K, Oelke M, Zeldis J. AIM™ platform: A new immunotherapy approach for viral diseases. Front Med (Lausanne) 2022; 9:1070529. [PMID: 36619639 PMCID: PMC9822776 DOI: 10.3389/fmed.2022.1070529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 12/07/2022] [Indexed: 12/25/2022] Open
Abstract
In addition to complications of acute diseases, chronic viral infections are linked to both malignancies and autoimmune disorders. Lack of adequate treatment options for Epstein-Barr virus (EBV), Human T-lymphotropic virus type 1 (HTLV-1), and human papillomavirus (HPV) remains. The NexImmune Artificial Immune Modulation (AIM) nanoparticle platform can be used to direct T cell responses by mimicking the dendritic cell function. In one application, AIM nanoparticles are used ex vivo to enrich and expand (E+E) rare populations of multi-antigen-specific CD8+ T cells for use of these cells as an AIM adoptive cell therapy. This study has demonstrated using E+E CD8+ T cells, the functional relevance of targeting EBV, HTLV-1, and HPV. Expanded T cells consist primarily of effector memory, central memory, and self-renewing stem-like memory T cells directed at selected viral antigen peptides presented by the AIM nanoparticle. T cells expanded against either EBV- or HPV-antigens were highly polyfunctional and displayed substantial in vitro cytotoxic activity against cell lines expressing the respective antigens. Our initial work was in the context of exploring T cells expanded from healthy donors and restricted to human leukocyte antigen (HLA)-A*02:01 serotype. AIM Adoptive Cell Therapies (ACT) are also being developed for other HLA class I serotypes. AIM adoptive cell therapies of autologous or allogeneic T cells specific to antigens associated with acute myeloid leukemia and multiple myeloma are currently in the clinic. The utility and flexibility of the AIM nanoparticle platform will be expanded as we advance the second application, an AIM injectable off-the-shelf nanoparticle, which targets multiple antigen-specific T cell populations to either activate, tolerize, or destroy these targeted CD8+ T cells directly in vivo, leaving non-target cells alone. The AIM injectable platform offers the potential to develop new multi-antigen specific therapies for treating infectious diseases, cancer, and autoimmune diseases.
Collapse
|
5
|
Marrella V, Facoetti A, Cassani B. Cellular Senescence in Immunity against Infections. Int J Mol Sci 2022; 23:11845. [PMID: 36233146 PMCID: PMC9570409 DOI: 10.3390/ijms231911845] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
Cellular senescence is characterized by irreversible cell cycle arrest in response to different triggers and an inflammatory secretome. Although originally described in fibroblasts and cell types of solid organs, cellular senescence affects most tissues with advancing age, including the lymphoid tissue, causing chronic inflammation and dysregulation of both innate and adaptive immune functions. Besides its normal occurrence, persistent microbial challenge or pathogenic microorganisms might also accelerate the activation of cellular aging, inducing the premature senescence of immune cells. Therapeutic strategies counteracting the detrimental effects of cellular senescence are being developed. Their application to target immune cells might have the potential to improve immune dysfunctions during aging and reduce the age-dependent susceptibility to infections. In this review, we discuss how immune senescence influences the host's ability to resolve more common infections in the elderly and detail the different markers proposed to identify such senescent cells; the mechanisms by which infectious agents increase the extent of immune senescence are also reviewed. Finally, available senescence therapeutics are discussed in the context of their effects on immunity and against infections.
Collapse
Affiliation(s)
- Veronica Marrella
- UOS Milan Unit, Istituto di Ricerca Genetica e Biomedica (IRGB), CNR, 20138 Milan, Italy
- IRCCS Humanitas Research Hospital, 20089 Milan, Italy
| | - Amanda Facoetti
- Department of Biomedical Sciences, Humanitas University, 20090 Milan, Italy
| | - Barbara Cassani
- IRCCS Humanitas Research Hospital, 20089 Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine, Università Degli Studi di Milano, 20089 Milan, Italy
| |
Collapse
|
6
|
Lammoglia Cobo MF, Ritter J, Gary R, Seitz V, Mautner J, Aigner M, Völkl S, Schaffer S, Moi S, Seegebarth A, Bruns H, Rösler W, Amann K, Büttner-Herold M, Hennig S, Mackensen A, Hummel M, Moosmann A, Gerbitz A. Reconstitution of EBV-directed T cell immunity by adoptive transfer of peptide-stimulated T cells in a patient after allogeneic stem cell transplantation for AITL. PLoS Pathog 2022; 18:e1010206. [PMID: 35452490 PMCID: PMC9067708 DOI: 10.1371/journal.ppat.1010206] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 05/04/2022] [Accepted: 03/31/2022] [Indexed: 11/29/2022] Open
Abstract
Reconstitution of the T cell repertoire after allogeneic stem cell transplantation is a long and often incomplete process. As a result, reactivation of Epstein-Barr virus (EBV) is a frequent complication that may be treated by adoptive transfer of donor-derived EBV-specific T cells. We generated donor-derived EBV-specific T cells by stimulation with peptides representing defined epitopes covering multiple HLA restrictions. T cells were adoptively transferred to a patient who had developed persisting high titers of EBV after allogeneic stem cell transplantation for angioimmunoblastic T-cell lymphoma (AITL). T cell receptor beta (TCRβ) deep sequencing showed that the T cell repertoire of the patient early after transplantation (day 60) was strongly reduced and only very low numbers of EBV-specific T cells were detectable. Manufacturing and in vitro expansion of donor-derived EBV-specific T cells resulted in enrichment of EBV epitope-specific, HLA-restricted T cells. Monitoring of T cell clonotypes at a molecular level after adoptive transfer revealed that the dominant TCR sequences from peptide-stimulated T cells persisted long-term and established an EBV-specific TCR clonotype repertoire in the host, with many of the EBV-specific TCRs present in the donor. This reconstituted repertoire was associated with immunological control of EBV and with lack of further AITL relapse. A characteristic feature of all herpesviruses is their persistence in the host’s body after primary infection. Hence, the host’s immune system is confronted with the problem to control these viruses life-long. When the immune system is severely compromised, for example after stem cell transplantation from a foreign (allogeneic) donor, these viruses can reappear, as they persist in the host’s body life-long after primary infection. Epstein-Barr virus (EBV) is a herpesvirus that can cause life-threatening complications after stem cell transplantation and only reinforcement of the host’s immune system can reestablish control over the virus. Here we show that ex vivo manufactured EBV-specific T cells can reestablish long-term control of EBV and that these cells persist in the host’s body over months. These results give us a better understanding of viral immune reconstitution post-transplant and of clinically-relevant T cell populations against EBV.
Collapse
Affiliation(s)
- María Fernanda Lammoglia Cobo
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Julia Ritter
- Institute of Pathology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Regina Gary
- Department of Internal Medicine 5 –Hematology/Oncology, University Hospital Erlangen, Erlangen, Germany
| | - Volkhard Seitz
- Institute of Pathology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- HS Diagnomics GmbH, Berlin, Germany
| | - Josef Mautner
- Department of Medicine III, LMU-Klinikum, Munich, Germany
- German Centre for Infection Research, Munich, Germany
| | - Michael Aigner
- Department of Internal Medicine 5 –Hematology/Oncology, University Hospital Erlangen, Erlangen, Germany
| | - Simon Völkl
- Department of Internal Medicine 5 –Hematology/Oncology, University Hospital Erlangen, Erlangen, Germany
| | - Stefanie Schaffer
- Department of Internal Medicine 5 –Hematology/Oncology, University Hospital Erlangen, Erlangen, Germany
| | - Stephanie Moi
- Department of Internal Medicine 5 –Hematology/Oncology, University Hospital Erlangen, Erlangen, Germany
| | - Anke Seegebarth
- Institute of Pathology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Heiko Bruns
- Department of Internal Medicine 5 –Hematology/Oncology, University Hospital Erlangen, Erlangen, Germany
| | - Wolf Rösler
- Department of Internal Medicine 5 –Hematology/Oncology, University Hospital Erlangen, Erlangen, Germany
| | - Kerstin Amann
- Department of Nephropathology, Institute of Pathology, University of Erlangen, Erlangen, Germany
| | - Maike Büttner-Herold
- Department of Nephropathology, Institute of Pathology, University of Erlangen, Erlangen, Germany
| | | | - Andreas Mackensen
- Department of Internal Medicine 5 –Hematology/Oncology, University Hospital Erlangen, Erlangen, Germany
| | - Michael Hummel
- Institute of Pathology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Andreas Moosmann
- Department of Medicine III, LMU-Klinikum, Munich, Germany
- German Centre for Infection Research, Munich, Germany
| | - Armin Gerbitz
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Center, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
7
|
Sooda A, Rwandamuriye F, Wanjalla CN, Jing L, Koelle DM, Peters B, Leary S, Chopra A, Calderwood MA, Mallal SA, Pavlos R, Watson M, Phillips EJ, Redwood AJ. Abacavir inhibits but does not cause self-reactivity to HLA-B*57:01-restricted EBV specific T cell receptors. Commun Biol 2022; 5:133. [PMID: 35173258 PMCID: PMC8850454 DOI: 10.1038/s42003-022-03058-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 01/14/2022] [Indexed: 01/11/2023] Open
Abstract
Pre-existing pathogen-specific memory T cell responses can contribute to multiple adverse outcomes including autoimmunity and drug hypersensitivity. How the specificity of the T cell receptor (TCR) is subverted or seconded in many of these diseases remains unclear. Here, we apply abacavir hypersensitivity (AHS) as a model to address this question because the disease is linked to memory T cell responses and the HLA risk allele, HLA-B*57:01, and the initiating insult, abacavir, are known. To investigate the role of pathogen-specific TCR specificity in mediating AHS we performed a genome-wide screen for HLA-B*57:01 restricted T cell responses to Epstein-Barr virus (EBV), one of the most prevalent human pathogens. T cell epitope mapping revealed HLA-B*57:01 restricted responses to 17 EBV open reading frames and identified an epitope encoded by EBNA3C. Using these data, we cloned the dominant TCR for EBNA3C and a previously defined epitope within EBNA3B. TCR specificity to each epitope was confirmed, however, cloned TCRs did not cross-react with abacavir plus self-peptide. Nevertheless, abacavir inhibited TCR interactions with their cognate ligands, demonstrating that TCR specificity may be subverted by a drug molecule. These results provide an experimental road map for future studies addressing the heterologous immune responses of TCRs including T cell mediated adverse drug reactions.
Collapse
Affiliation(s)
- Anuradha Sooda
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, WA, Australia
| | - Francois Rwandamuriye
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, WA, Australia
- Telethon Kids Institute, Nedlands, WA, Australia
| | - Celestine N Wanjalla
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lichen Jing
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - David M Koelle
- Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
- Benaroya Research Institute, Seattle, WA, USA
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Bjoern Peters
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Shay Leary
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, WA, Australia
| | - Abha Chopra
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, WA, Australia
| | - Michael A Calderwood
- Department of Medicine, The Channing Laboratory, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Simon A Mallal
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, WA, Australia
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rebecca Pavlos
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, WA, Australia
- Telethon Kids Institute, Nedlands, WA, Australia
| | - Mark Watson
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, WA, Australia
| | - Elizabeth J Phillips
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, WA, Australia.
- Center for Drug Safety & Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Alec J Redwood
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, WA, Australia
- Institute for Respiratory Health, Level 2, 6 Verdun Street, Nedlands, WA, 6009, Australia
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
8
|
Clark F, Gil A, Thapa I, Aslan N, Ghersi D, Selin LK. Cross-reactivity influences changes in human influenza A virus and Epstein Barr virus specific CD8 memory T cell receptor alpha and beta repertoires between young and old. Front Immunol 2022; 13:1011935. [PMID: 36923729 PMCID: PMC10009332 DOI: 10.3389/fimmu.2022.1011935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 12/30/2022] [Indexed: 03/03/2023] Open
Abstract
Older people have difficulty controlling infection with common viruses such as influenza A virus (IAV), RNA virus which causes recurrent infections due to a high rate of genetic mutation, and Epstein Barr virus (EBV), DNA virus which persists in B cells for life in the 95% of people that become acutely infected. We questioned whether changes in epitope-specific memory CD8 T cell receptor (TCR) repertoires to these two common viruses could occur with increasing age and contribute to waning immunity. We compared CD8 memory TCR alpha and beta repertoires in two HLA-A2+ EBV- and IAV-immune donors, young (Y) and older (O) donors to three immunodominant epitopes known to be cross-reactive, IAV-M158-66 (IAV-M1), EBV-BMLF1280-288 (EBV-BM), and EBV-BRLF1109-117 (EBV-BR). We, therefore, also designed these studies to examine if TCR cross-reactivity could contribute to changes in repertoire with increasing age. TCR high throughput sequencing showed a significant difference in the pattern of TRBV usage between Y and O. However, there were many more differences in AV and AJ usage, between the age groups suggesting that changes in TCRα usage may play a greater role in evolution of the TCR repertoire emphasizing the importance of studying TRAV repertoires. With increasing age there was a preferential retention of TCR for all three epitopes with features in their complementarity-determining region (CDR3) that increased their ease of generation, and their cross-reactive potential. Young and older donors differed in the patterns of AV/AJ and BV/BJ pairings and usage of dominant CDR3 motifs specific to all three epitopes. Both young and older donors had cross-reactive responses between these 3 epitopes, which were unique and differed from the cognate responses having features that suggested they could interact with either ligand. There was an increased tendency for the classic IAV-M1 specific clone BV19-IRSS-JB2.7/AV27-CAGGGSQGNLIF-AJ42 to appear among the cross-reactive clones, suggesting that the dominance of this clone may relate to its cross-reactivity with EBV. These results suggest that although young and older donors retain classic TCR features for each epitope their repertoires are gradually changing with age, maintaining TCRs that are cross-reactive between these two common human viruses, one with recurrent infections and the other a persistent virus which frequently reactivates.
Collapse
Affiliation(s)
- Fransenio Clark
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Anna Gil
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Ishwor Thapa
- School of Interdisciplinary Informatics, University of Nebraska at Omaha, Omaha, NE, United States
| | - Nuray Aslan
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Dario Ghersi
- School of Interdisciplinary Informatics, University of Nebraska at Omaha, Omaha, NE, United States
| | - Liisa K Selin
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
9
|
Lanfermeijer J, Nühn MM, Emmelot ME, Poelen MCM, van Els CACM, Borghans JAM, van Baarle D, Kaaijk P, de Wit J. Longitudinal Characterization of the Mumps-Specific HLA-A2 Restricted T-Cell Response after Mumps Virus Infection. Vaccines (Basel) 2021; 9:1431. [PMID: 34960178 PMCID: PMC8707000 DOI: 10.3390/vaccines9121431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/19/2021] [Accepted: 11/30/2021] [Indexed: 11/16/2022] Open
Abstract
Waning of the mumps virus (MuV)-specific humoral response after vaccination has been suggested as a cause for recent mumps outbreaks in vaccinated young adults, although it cannot explain all cases. Moreover, CD8+ T cells may play an important role in the response against MuV; however, little is known about the characteristics and dynamics of the MuV-specific CD8+ T-cell response after MuV infection. Here, we had the opportunity to follow the CD8+ T-cell response to three recently identified HLA-A2*02:01-restricted MuV-specific epitopes from 1.5 to 36 months post-MuV infection in five previously vaccinated and three unvaccinated individuals. The infection-induced CD8+ T-cell response was dominated by T cells specific for the ALDQTDIRV and LLDSSTTRV epitopes, while the response to the GLMEGQIVSV epitope was subdominant. MuV-specific CD8+ T-cell frequencies in the blood declined between 1.5 and 9 months after infection. This decline was not explained by changes in the expression of inhibitory receptors or homing markers. Despite the ongoing changes in the frequencies and phenotype of MuV-specific CD8+ T cells, TCRβ analyses revealed a stable MuV-specific T-cell repertoire over time. These insights in the maintenance of the cellular response against mumps may provide hallmarks for optimizing vaccination strategies towards a long-term cellular memory response.
Collapse
Affiliation(s)
- Josien Lanfermeijer
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands; (J.L.); (M.M.N.); (M.E.E.); (M.C.M.P.); (C.A.C.M.v.E.); (D.v.B.); (P.K.)
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands;
| | - Marieke M. Nühn
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands; (J.L.); (M.M.N.); (M.E.E.); (M.C.M.P.); (C.A.C.M.v.E.); (D.v.B.); (P.K.)
| | - Maarten E. Emmelot
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands; (J.L.); (M.M.N.); (M.E.E.); (M.C.M.P.); (C.A.C.M.v.E.); (D.v.B.); (P.K.)
| | - Martien C. M. Poelen
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands; (J.L.); (M.M.N.); (M.E.E.); (M.C.M.P.); (C.A.C.M.v.E.); (D.v.B.); (P.K.)
| | - Cécile A. C. M. van Els
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands; (J.L.); (M.M.N.); (M.E.E.); (M.C.M.P.); (C.A.C.M.v.E.); (D.v.B.); (P.K.)
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - José A. M. Borghans
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands;
| | - Debbie van Baarle
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands; (J.L.); (M.M.N.); (M.E.E.); (M.C.M.P.); (C.A.C.M.v.E.); (D.v.B.); (P.K.)
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands;
| | - Patricia Kaaijk
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands; (J.L.); (M.M.N.); (M.E.E.); (M.C.M.P.); (C.A.C.M.v.E.); (D.v.B.); (P.K.)
| | - Jelle de Wit
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, 3721 MA Bilthoven, The Netherlands; (J.L.); (M.M.N.); (M.E.E.); (M.C.M.P.); (C.A.C.M.v.E.); (D.v.B.); (P.K.)
| |
Collapse
|
10
|
Mazouz S, Boisvert M, Abdel-Hakeem MS, Khedr O, Bruneau J, Shoukry NH. Expansion of Unique Hepatitis C Virus-Specific Public CD8 + T Cell Clonotypes during Acute Infection and Reinfection. THE JOURNAL OF IMMUNOLOGY 2021; 207:1180-1193. [PMID: 34341170 DOI: 10.4049/jimmunol.2001386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 06/09/2021] [Indexed: 11/19/2022]
Abstract
Hepatitis C virus (HCV) infection resolves spontaneously in ∼25% of acutely infected humans where viral clearance is mediated primarily by virus-specific CD8+ T cells. Previous cross-sectional analysis of the CD8+ TCR repertoire targeting two immunodominant HCV epitopes reported widespread use of public TCRs shared by different subjects, irrespective of infection outcome. However, little is known about the evolution of the public TCR repertoire during acute HCV and whether cross-reactivity to other Ags can influence infectious outcome. In this article, we analyzed the CD8+ TCR repertoire specific to the immunodominant and cross-reactive HLA-A2-restricted nonstructural 3-1073 epitope during acute HCV in humans progressing to either spontaneous resolution or chronic infection and at ∼1 y after viral clearance. TCR repertoire diversity was comparable among all groups with preferential usage of the TCR-β V04 and V06 gene families. We identified a set of 13 public clonotypes in HCV-infected humans independent of infection outcome. Six public clonotypes used the V04 gene family. Several public clonotypes were long-lived in resolvers and expanded on reinfection. By mining publicly available data, we identified several low-frequency CDR3 sequences in the HCV-specific repertoire matching human TCRs specific for other HLA-A2-restricted epitopes from melanoma, CMV, influenza A, EBV, and yellow fever viruses, but they were of low frequency and limited cross-reactivity. In conclusion, we identified 13 new public human CD8+ TCR clonotypes unique to HCV that expanded during acute infection and reinfection. The low frequency of cross-reactive TCRs suggests that they are not major determinants of infectious outcome.
Collapse
Affiliation(s)
- Sabrina Mazouz
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université de Montreal, Montreal, Quebec, Canada
| | - Maude Boisvert
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Mohamed S Abdel-Hakeem
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université de Montreal, Montreal, Quebec, Canada
| | - Omar Khedr
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Julie Bruneau
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada.,Département de Médecine Familiale et de Médecine d'Urgence, Université de Montréal, Montreal, Quebec, Canada; and
| | - Naglaa H Shoukry
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada; .,Département de Médecine, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
11
|
Fernandes SG, Dsouza R, Khattar E. External environmental agents influence telomere length and telomerase activity by modulating internal cellular processes: Implications in human aging. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 85:103633. [PMID: 33711516 DOI: 10.1016/j.etap.2021.103633] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 01/30/2021] [Accepted: 03/05/2021] [Indexed: 06/12/2023]
Abstract
External environment affects cellular physiological processes and impact the stability of our genome. The most important structural components of our linear chromosomes which endure the impact by these agents, are the chromosomal ends called telomeres. Telomeres preserve the integrity of our genome by preventing end to end fusions and telomeric loss through by inhibiting DNA damage response (DDR) activation. This is accomplished by the presence of a six membered shelterin complex at telomeres. Further, telomeres cannot be replicated by normal DNA polymerase and require a special enzyme called telomerase which is expressed only in stem cells, few immune cells and germ cells. Telomeres are rich in guanine content and thus become extremely prone to damage arising due to physiological processes like oxidative stress and inflammation. External environmental factors which includes various physical, biological and chemical agents also affect telomere homeostasis by increasing oxidative stress and inflammation. In the present review, we highlight the effect of these external factors on telomerase activity and telomere length. We also discuss how the external agents affect the physiological processes, thus modulating telomere stability. Further, we describe its implication in the development of aging and its related pathologies.
Collapse
Affiliation(s)
- Stina George Fernandes
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be University), Vile Parle West, Mumbai, 400056, India
| | - Rebecca Dsouza
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be University), Vile Parle West, Mumbai, 400056, India
| | - Ekta Khattar
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be University), Vile Parle West, Mumbai, 400056, India.
| |
Collapse
|
12
|
Lanfermeijer J, de Greef PC, Hendriks M, Vos M, van Beek J, Borghans JAM, van Baarle D. Age and CMV-Infection Jointly Affect the EBV-Specific CD8 + T-Cell Repertoire. FRONTIERS IN AGING 2021; 2:665637. [PMID: 35822032 PMCID: PMC9261403 DOI: 10.3389/fragi.2021.665637] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 03/31/2021] [Indexed: 01/15/2023]
Abstract
CD8+ T cells play an important role in protection against viral infections. With age, changes in the T-cell pool occur, leading to diminished responses against both new and recurring infections in older adults. This is thought to be due to a decrease in both T-cell numbers and T-cell receptor (TCR) diversity. Latent infection with cytomegalovirus (CMV) is assumed to contribute to this age-associated decline of the immune system. The observation that the level of TCR diversity in the total memory T-cell pool stays relatively stable during aging is remarkable in light of the constant input of new antigen-specific memory T cells. What happens with the diversity of the individual antigen-specific T-cell repertoires in the memory pool remains largely unknown. Here we studied the effect of aging on the phenotype and repertoire diversity of CMV-specific and Epstein-Barr virus (EBV)-specific CD8+ T cells, as well as the separate effects of aging and CMV-infection on the EBV-specific T-cell repertoire. Antigen-specific T cells against both persistent viruses showed an age-related increase in the expression of markers associated with a more differentiated phenotype, including KLRG-1, an increase in the fraction of terminally differentiated T cells, and a decrease in the diversity of the T-cell repertoire. Not only age, but also CMV infection was associated with a decreased diversity of the EBV-specific T-cell repertoire. This suggests that both CMV infection and age can impact the T-cell repertoire against other antigens.
Collapse
Affiliation(s)
- Josien Lanfermeijer
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Peter C. de Greef
- Theoretical Biology and Bioinformatics, Utrecht University, Utrecht, Netherlands
| | - Marion Hendriks
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Martijn Vos
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Josine van Beek
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - José A. M. Borghans
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Debbie van Baarle
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
13
|
CD4 + T Cells Recognize Conserved Influenza A Epitopes through Shared Patterns of V-Gene Usage and Complementary Biochemical Features. Cell Rep 2021; 32:107885. [PMID: 32668259 PMCID: PMC7370177 DOI: 10.1016/j.celrep.2020.107885] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 01/20/2020] [Accepted: 06/17/2020] [Indexed: 12/15/2022] Open
Abstract
T cell recognition of peptides presented by human leukocyte antigens (HLAs) is mediated by the highly variable T cell receptor (TCR). Despite this built-in TCR variability, individuals can mount immune responses against viral epitopes by using identical or highly related TCRs expressed on CD8+ T cells. Characterization of these TCRs has extended our understanding of the molecular mechanisms that govern the recognition of peptide-HLA. However, few examples exist for CD4+ T cells. Here, we investigate CD4+ T cell responses to the internal proteins of the influenza A virus that correlate with protective immunity. We identify five internal epitopes that are commonly recognized by CD4+ T cells in five HLA-DR1+ subjects and show conservation across viral strains and zoonotic reservoirs. TCR repertoire analysis demonstrates several shared gene usage biases underpinned by complementary biochemical features evident in a structural comparison. These epitopes are attractive targets for vaccination and other T cell therapies.
Collapse
|
14
|
Lanfermeijer J, Borghans JAM, Baarle D. How age and infection history shape the antigen-specific CD8 + T-cell repertoire: Implications for vaccination strategies in older adults. Aging Cell 2020; 19:e13262. [PMID: 33078890 PMCID: PMC7681067 DOI: 10.1111/acel.13262] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/21/2020] [Accepted: 09/25/2020] [Indexed: 12/21/2022] Open
Abstract
Older adults often show signs of impaired CD8+ T‐cell immunity, reflected by weaker responses against new infections and vaccinations, and decreased protection against reinfection. This immune impairment is in part thought to be the consequence of a decrease in both T‐cell numbers and repertoire diversity. If this is indeed the case, a strategy to prevent infectious diseases in older adults could be the induction of protective memory responses through vaccination at a younger age. However, this requires that the induced immune responses are maintained until old age. It is therefore important to obtain insights into the long‐term maintenance of the antigen‐specific T‐cell repertoire. Here, we review the literature on the maintenance of antigen‐experienced CD8+ T‐cell repertoires against acute and chronic infections. We describe the complex interactions that play a role in shaping the memory T‐cell repertoire, and the effects of age, infection history, and T‐cell avidity. We discuss the implications of these findings for the development of new vaccination strategies to protect older adults.
Collapse
Affiliation(s)
- Josien Lanfermeijer
- Center for Infectious Disease Control National Institute for Public Health and the Environment Bilthoven the Netherlands
- Center for Translational Immunology University Medical Center Utrecht the Netherlands
| | - José A. M. Borghans
- Center for Translational Immunology University Medical Center Utrecht the Netherlands
| | - Debbie Baarle
- Center for Infectious Disease Control National Institute for Public Health and the Environment Bilthoven the Netherlands
- Center for Translational Immunology University Medical Center Utrecht the Netherlands
- Virology & Immunology Research Department of Medical Microbiology and Infection prevention University Medical Center Groningen the Netherlands
| |
Collapse
|
15
|
T Cell Receptor Diversity and Lineage Relationship between Virus-Specific CD8 T Cell Subsets during Chronic Lymphocytic Choriomeningitis Virus Infection. J Virol 2020; 94:JVI.00935-20. [PMID: 32759317 DOI: 10.1128/jvi.00935-20] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 07/24/2020] [Indexed: 01/08/2023] Open
Abstract
Recent studies on chronic viral infections have defined a novel programmed cell death 1-positive (PD-1+) T cell factor 1-positive (TCF1+) stem-like CD8 T cell subset that gives rise to the terminally differentiated exhausted CD8 T cells. In this study, we performed T cell receptor beta (TCRβ) sequencing of virus-specific CD8 T cells during chronic lymphocytic choriomeningitis virus (LCMV) infection to examine the TCR diversity and lineage relationship of these two functionally distinct subsets. We found that >95% of the TCR repertoire of the exhausted CD8 T cell subset was shared with the stem-like CD8 T cells. The TCR repertoires of both CD8 T cell subsets were composed mostly of a few dominant clonotypes, but there was slightly more breadth and diversity in the stem-like CD8 T cells than their exhausted counterpart (∼40 versus ∼15 GP33+ clonotypes; ∼20 versus ∼7 GP276+ clonotypes). Interestingly, the breadth of the TCR repertoire was broader during the early stages (day 8) of the chronic infection than the later stages (days 45 to 60), showing that there was a narrowing of the TCR repertoire during chronic infection (∼2-fold GP33+ and GP276+ stem-like subset; ∼10-fold GP33+ and ∼5-fold GP276+ exhausted subset). In contrast, during acute LCMV infection, the TCR repertoire was much broader in both GP33-specific effector (∼160 clonotypes) and memory CD8 T cells (∼160 clonotypes). Overall, our data demonstrate that the virus-specific CD8 T cell TCR repertoire is broad and remains stable after acute LCMV infection, but it contracts and is narrower during chronic infection. Our study also shows that the repertoire of the exhausted CD8 T cell subset is almost completely derived from the stem-like CD8 T cell subset during established chronic LCMV infection.IMPORTANCE CD8 TCR repertoires responding to chronic viral infections (HIV, hepatitis C virus [HCV], Epstein-Barr virus [EBV], and cytomegalovirus [CMV]) have limited breadth and diversity. How these repertoires change and are maintained throughout the chronic infection are unknown. We thus characterized the LCMV-specific CD8 TCR repertoires of stem-like and terminally exhausted subsets generated during chronic LCMV infections. During chronic LCMV infections, the repertoires started as diverse but became more clonal at the late time point. Further, the exhausted subset was composed of dominant clonotypes that were shared with the stem-like subset. Together, we demonstrate that the TCR repertoire contracts over time and is almost exclusively derived from the stem-like subset late during the persistent viral infection. Our data suggest that dominant clonotypes in the exhausted subset are derived from a diverse pool of stem-like clonotypes, which may be contributing to the clonality observed during chronic viral infections.
Collapse
|
16
|
Rowntree LC, Nguyen THO, Farenc C, Halim H, Hensen L, Rossjohn J, Kotsimbos TC, Purcell AW, Kedzierska K, Gras S, Mifsud NA. A Shared TCR Bias toward an Immunogenic EBV Epitope Dominates in HLA-B*07:02–Expressing Individuals. THE JOURNAL OF IMMUNOLOGY 2020; 205:1524-1534. [DOI: 10.4049/jimmunol.2000249] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/19/2020] [Indexed: 11/19/2022]
|
17
|
Gil A, Kamga L, Chirravuri-Venkata R, Aslan N, Clark F, Ghersi D, Luzuriaga K, Selin LK. Epstein-Barr Virus Epitope-Major Histocompatibility Complex Interaction Combined with Convergent Recombination Drives Selection of Diverse T Cell Receptor α and β Repertoires. mBio 2020; 11:e00250-20. [PMID: 32184241 PMCID: PMC7078470 DOI: 10.1128/mbio.00250-20] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 02/11/2020] [Indexed: 01/07/2023] Open
Abstract
Recognition modes of individual T cell receptors (TCRs) are well studied, but factors driving the selection of TCR repertoires from primary through persistent human virus infections are less well understood. Using deep sequencing, we demonstrate a high degree of diversity of Epstein-Barr virus (EBV)-specific clonotypes in acute infectious mononucleosis (AIM). Only 9% of unique clonotypes detected in AIM persisted into convalescence; the majority (91%) of unique clonotypes detected in AIM were not detected in convalescence and were seeming replaced by equally diverse "de novo" clonotypes. The persistent clonotypes had a greater probability of being generated than nonpersistent clonotypes due to convergence recombination of multiple nucleotide sequences to encode the same amino acid sequence, as well as the use of shorter complementarity-determining regions 3 (CDR3s) with fewer nucleotide additions (i.e., sequences closer to germ line). Moreover, the two most immunodominant HLA-A2-restricted EBV epitopes, BRLF1109 and BMLF1280, show highly distinct antigen-specific public (i.e., shared between individuals) features. In fact, TCRα CDR3 motifs played a dominant role, while TCRβ played a minimal role, in the selection of TCR repertoire to an immunodominant EBV epitope, BRLF1. This contrasts with the majority of previously reported repertoires, which appear to be selected either on TCRβ CDR3 interactions with peptide/major histocompatibility complex (MHC) or in combination with TCRα CDR3. Understanding of how TCR-peptide-MHC complex interactions drive repertoire selection can be used to develop optimal strategies for vaccine design or generation of appropriate adoptive immunotherapies for viral infections in transplant settings or for cancer.IMPORTANCE Several lines of evidence suggest that TCRα and TCRβ repertoires play a role in disease outcomes and treatment strategies during viral infections in transplant patients and in cancer and autoimmune disease therapy. Our data suggest that it is essential that we understand the basic principles of how to drive optimum repertoires for both TCR chains, α and β. We address this important issue by characterizing the CD8 TCR repertoire to a common persistent human viral infection (EBV), which is controlled by appropriate CD8 T cell responses. The ultimate goal would be to determine if the individuals who are infected asymptomatically develop a different TCR repertoire than those that develop the immunopathology of AIM. Here, we begin by doing an in-depth characterization of both CD8 T cell TCRα and TCRβ repertoires to two immunodominant EBV epitopes over the course of AIM, identifying potential factors that may be driving their selection.
Collapse
Affiliation(s)
- Anna Gil
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Larisa Kamga
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | | - Nuray Aslan
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Fransenio Clark
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Dario Ghersi
- School of Interdisciplinary Informatics, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Katherine Luzuriaga
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Liisa K Selin
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
18
|
Kamga L, Gil A, Song I, Brody R, Ghersi D, Aslan N, Stern LJ, Selin LK, Luzuriaga K. CDR3α drives selection of the immunodominant Epstein Barr virus (EBV) BRLF1-specific CD8 T cell receptor repertoire in primary infection. PLoS Pathog 2019; 15:e1008122. [PMID: 31765434 PMCID: PMC6901265 DOI: 10.1371/journal.ppat.1008122] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 12/09/2019] [Accepted: 10/03/2019] [Indexed: 12/20/2022] Open
Abstract
The T cell receptor (TCR) repertoire is an essential component of the CD8 T-cell immune response. Here, we seek to investigate factors that drive selection of TCR repertoires specific to the HLA-A2-restricted immunodominant epitope BRLF1109-117 (YVLDHLIVV) over the course of primary Epstein Barr virus (EBV) infection. Using single-cell paired TCRαβ sequencing of tetramer sorted CD8 T cells ex vivo, we show at the clonal level that recognition of the HLA-A2-restricted BRLF1 (YVL-BR, BRLF-1109) epitope is mainly driven by the TCRα chain. For the first time, we identify a CDR3α (complementarity determining region 3 α) motif, KDTDKL, resulting from an obligate AV8.1-AJ34 pairing that was shared by all four individuals studied. This observation coupled with the fact that this public AV8.1-KDTDKL-AJ34 TCR pairs with multiple different TCRβ chains within the same donor (median 4; range: 1–9), suggests that there are some unique structural features of the interaction between the YVL-BR/MHC and the AV8.1-KDTDKL-AJ34 TCR that leads to this high level of selection. Newly developed TCR motif algorithms identified a lysine at position 1 of the CDR3α motif that is highly conserved and likely important for antigen recognition. Crystal structure analysis of the YVL-BR/HLA-A2 complex revealed that the MHC-bound peptide bulges at position 4, exposing a negatively charged aspartic acid that may interact with the positively charged lysine of CDR3α. TCR cloning and site-directed mutagenesis of the CDR3α lysine ablated YVL-BR-tetramer staining and substantially reduced CD69 upregulation on TCR mutant-transduced cells following antigen-specific stimulation. Reduced activation of T cells expressing this CDR3 motif was also observed following exposure to mutated (D4A) peptide. In summary, we show that a highly public TCR repertoire to an immunodominant epitope of a common human virus is almost completely selected on the basis of CDR3α and provide a likely structural basis for the selection. These studies emphasize the importance of examining TCRα, as well as TCRβ, in understanding the CD8 T cell receptor repertoire. EBV is a ubiquitous human virus that has been linked to several diseases, including cancers and post-transplant lymphoproliferative disorders. CD8 T cells are important for controlling EBV replication. Generation and maintenance of virus-specific CD8 T cells is dependent on specific interaction between MHC-peptide complexes on the infected cell and the TCR. In this study, we performed single cell sequencing of paired TCR α and β chains from EBV-specific CD8 T cells isolated at two time points (primary infection and convalescence) from four individuals undergoing acute EBV infection. We describe a TCRα sequence that was shared by all four individuals and identify conserved residues within this sequence that likely contribute to viral recognition. Examination of the crystal structure of the peptide-MHC complex and subsequent experimental data suggest that a specific interaction between a negatively charged aspartic acid at position 4 of the peptide and a positively charged lysine in the TCR may be particularly important. These findings are highly relevant to current efforts to understand how the TCR repertoire may contribute to or protect against disease, the development of TCR diagnostics for diseases, and at improving the efficacy of T cell based therapies.
Collapse
MESH Headings
- Amino Acid Sequence
- CD8-Positive T-Lymphocytes/immunology
- Complementarity Determining Regions/genetics
- Complementarity Determining Regions/immunology
- Complementarity Determining Regions/metabolism
- Epitopes, T-Lymphocyte/immunology
- Epstein-Barr Virus Infections/immunology
- Epstein-Barr Virus Infections/virology
- HLA-A2 Antigen/immunology
- Herpesvirus 4, Human/immunology
- Humans
- Immediate-Early Proteins/genetics
- Immediate-Early Proteins/immunology
- Immediate-Early Proteins/metabolism
- Immunodominant Epitopes/immunology
- Peptide Fragments/immunology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- T-Lymphocytes, Cytotoxic/immunology
- Trans-Activators/genetics
- Trans-Activators/immunology
- Trans-Activators/metabolism
Collapse
Affiliation(s)
- Larisa Kamga
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Anna Gil
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Inyoung Song
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Robin Brody
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Dario Ghersi
- School of Interdisciplinary Informatics, University of Nebraska at Omaha, Nebraska, United States of America
| | - Nuray Aslan
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Lawrence J. Stern
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Liisa K. Selin
- Department of Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail: (LKS); (KL)
| | - Katherine Luzuriaga
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail: (LKS); (KL)
| |
Collapse
|
19
|
Johnson DB, McDonnell WJ, Gonzalez-Ericsson PI, Al-Rohil RN, Mobley BC, Salem JE, Wang DY, Sanchez V, Wang Y, Chastain CA, Barker K, Liang Y, Warren S, Beechem JM, Menzies AM, Tio M, Long GV, Cohen JV, Guidon AC, O'Hare M, Chandra S, Chowdhary A, Lebrun-Vignes B, Goldinger SM, Rushing EJ, Buchbinder EI, Mallal SA, Shi C, Xu Y, Moslehi JJ, Sanders ME, Sosman JA, Balko JM. A case report of clonal EBV-like memory CD4 + T cell activation in fatal checkpoint inhibitor-induced encephalitis. Nat Med 2019; 25:1243-1250. [PMID: 31332390 PMCID: PMC6689251 DOI: 10.1038/s41591-019-0523-2] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 06/12/2019] [Indexed: 12/18/2022]
Abstract
Checkpoint inhibitors produce durable responses in numerous metastatic cancers, but immune-related adverse events (irAEs) complicate and limit their benefit. IrAEs can affect organ systems idiosyncratically; presentations range from mild and self-limited to fulminant and fatal. The molecular mechanisms underlying irAEs are poorly understood. Here, we report a fatal case of encephalitis arising during anti-programmed cell death receptor 1 therapy in a patient with metastatic melanoma. Histologic analyses revealed robust T cell infiltration and prominent programmed death ligand 1 expression. We identified 209 reported cases in global pharmacovigilance databases (across multiple cancer types) of encephalitis associated with checkpoint inhibitor regimens, with a 19% fatality rate. We performed further analyses from the index case and two additional cases to shed light on this recurrent and fulminant irAE. Spatial and multi-omic analyses pinpointed activated memory CD4+ T cells as highly enriched in the inflamed, affected region. We identified a highly oligoclonal T cell receptor repertoire, which we localized to activated memory cytotoxic (CD45RO+GZMB+Ki67+) CD4 cells. We also identified Epstein-Barr virus-specific T cell receptors and EBV+ lymphocytes in the affected region, which we speculate contributed to neural inflammation in the index case. Collectively, the three cases studied here identify CD4+ and CD8+ T cells as culprits of checkpoint inhibitor-associated immune encephalitis.
Collapse
Affiliation(s)
- Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA. .,Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Wyatt J McDonnell
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.,Center for Translational Immunology and Infectious Disease, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Rami N Al-Rohil
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Pathology and Dermatology, Duke University Medical Center, Durham, NC, USA
| | - Bret C Mobley
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joe-Elie Salem
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Sorbonne Université, INSERM CIC Paris-Est, AP-HP, ICAN, Regional Pharmacovigilance Centre, Pitié-Salpêtrière Hospital, Department of Pharmacology, Paris, France
| | - Daniel Y Wang
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Violeta Sanchez
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yu Wang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cody A Chastain
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Yan Liang
- NanoString Technologies, Seattle, WA, USA
| | | | | | - Alexander M Menzies
- Melanoma Institute Australia, Sydney, Australia.,The University of Sydney, Sydney, New South Wales, Australia.,Royal North Shore Hospital, Sydney, New South Wales, Australia.,Mater Hospital, Sydney, New South Wales, Australia
| | - Martin Tio
- Melanoma Institute Australia, Sydney, Australia
| | - Georgina V Long
- Melanoma Institute Australia, Sydney, Australia.,The University of Sydney, Sydney, New South Wales, Australia.,Royal North Shore Hospital, Sydney, New South Wales, Australia.,Mater Hospital, Sydney, New South Wales, Australia
| | | | | | | | - Sunandana Chandra
- Feinberg School of Medicine, Northwestern University, Evanston, IL, USA
| | - Akansha Chowdhary
- Feinberg School of Medicine, Northwestern University, Evanston, IL, USA
| | - Bénédicte Lebrun-Vignes
- Sorbonne Université, INSERM CIC Paris-Est, AP-HP, ICAN, Regional Pharmacovigilance Centre, Pitié-Salpêtrière Hospital, Department of Pharmacology, Paris, France
| | | | | | | | - Simon A Mallal
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.,Center for Translational Immunology and Infectious Disease, Vanderbilt University Medical Center, Nashville, TN, USA.,Institute for Immunology and Infectious Diseases, Perth, Australia
| | - Chanjuan Shi
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yaomin Xu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Javid J Moslehi
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Melinda E Sanders
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Justin M Balko
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA. .,Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA. .,Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA. .,Breast Cancer Research Program, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
20
|
Meckiff BJ, Ladell K, McLaren JE, Ryan GB, Leese AM, James EA, Price DA, Long HM. Primary EBV Infection Induces an Acute Wave of Activated Antigen-Specific Cytotoxic CD4 + T Cells. THE JOURNAL OF IMMUNOLOGY 2019; 203:1276-1287. [PMID: 31308093 PMCID: PMC6697742 DOI: 10.4049/jimmunol.1900377] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 06/20/2019] [Indexed: 12/14/2022]
Abstract
Primary EBV infection drives highly cytotoxic virus-specific CD4+ T cell responses. EBV-specific memory CD4+ T cells are polyfunctional but lack cytotoxic activity. Acute EBV-specific CD4-CTLs differ transcriptionally from classical memory CD4-CTLs.
CD4+ T cells are essential for immune protection against viruses, yet their multiple roles remain ill-defined at the single-cell level in humans. Using HLA class II tetramers, we studied the functional properties and clonotypic architecture of EBV-specific CD4+ T cells in patients with infectious mononucleosis, a symptomatic manifestation of primary EBV infection, and in long-term healthy carriers of EBV. We found that primary infection elicited oligoclonal expansions of TH1-like EBV-specific CD4+ T cells armed with cytotoxic proteins that responded immediately ex vivo to challenge with EBV-infected B cells. Importantly, these acutely generated cytotoxic CD4+ T cells were highly activated and transcriptionally distinct from classically described cytotoxic CD4+ memory T cells that accumulate during other persistent viral infections, including CMV and HIV. In contrast, EBV-specific memory CD4+ T cells displayed increased cytokine polyfunctionality but lacked cytotoxic activity. These findings suggested an important effector role for acutely generated cytotoxic CD4+ T cells that could potentially be harnessed to improve the efficacy of vaccines against EBV.
Collapse
Affiliation(s)
- Benjamin J Meckiff
- Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Kristin Ladell
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom; and
| | - James E McLaren
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom; and
| | - Gordon B Ryan
- Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Alison M Leese
- Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Eddie A James
- Tetramer Core Laboratory, Diabetes Program, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101
| | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom; and
| | - Heather M Long
- Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom;
| |
Collapse
|
21
|
Chicaybam L, Abdo L, Carneiro M, Peixoto B, Viegas M, de Sousa P, Fornazin MC, Spago MC, Albertoni Laranjeira AB, de Campos-Lima PO, Nowill A, Barros LRC, Bonamino MH. CAR T Cells Generated UsingSleeping BeautyTransposon Vectors and Expanded with an EBV-Transformed Lymphoblastoid Cell Line Display Antitumor ActivityIn VitroandIn Vivo. Hum Gene Ther 2019; 30:511-522. [DOI: 10.1089/hum.2018.218] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Leonardo Chicaybam
- Molecular Carcinogenesis Program, National Cancer Institute (INCA), Rio de Janeiro, Brazil
- Vice-Presidency of Research and Biological Collections, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Luiza Abdo
- Molecular Carcinogenesis Program, National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Mayra Carneiro
- Molecular Carcinogenesis Program, National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Bárbara Peixoto
- Cell Biology Program, National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Mariana Viegas
- Molecular Carcinogenesis Program, National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Priscila de Sousa
- Molecular Carcinogenesis Program, National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Márcia C. Fornazin
- Integrated Center for Oncohematology Research in Infancy, Institute of Biology, University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | - Maria C. Spago
- Integrated Center for Oncohematology Research in Infancy, Institute of Biology, University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | | | - Pedro O. de Campos-Lima
- Institute of Molecular and Cellular Engineering, Boldrini Children's Center, Campinas, Sao Paulo, Brazil
- Functional and Molecular Biology Program, Institute of Biology, University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | - Alexandre Nowill
- Integrated Center for Oncohematology Research in Infancy, Institute of Biology, University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | | | - Martín H. Bonamino
- Molecular Carcinogenesis Program, National Cancer Institute (INCA), Rio de Janeiro, Brazil
- Vice-Presidency of Research and Biological Collections, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
22
|
Bellon M, Nicot C. Telomere Dynamics in Immune Senescence and Exhaustion Triggered by Chronic Viral Infection. Viruses 2017; 9:v9100289. [PMID: 28981470 PMCID: PMC5691640 DOI: 10.3390/v9100289] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 09/28/2017] [Accepted: 09/29/2017] [Indexed: 02/06/2023] Open
Abstract
The progressive loss of immunological memory during aging correlates with a reduced proliferative capacity and shortened telomeres of T cells. Growing evidence suggests that this phenotype is recapitulated during chronic viral infection. The antigenic volume imposed by persistent and latent viruses exposes the immune system to unique challenges that lead to host T-cell exhaustion, characterized by impaired T-cell functions. These dysfunctional memory T cells lack telomerase, the protein capable of extending and stabilizing chromosome ends, imposing constraints on telomere dynamics. A deleterious consequence of this excessive telomere shortening is the premature induction of replicative senescence of viral-specific CD8+ memory T cells. While senescent cells are unable to expand, they can survive for extended periods of time and are more resistant to apoptotic signals. This review takes a closer look at T-cell exhaustion in chronic viruses known to cause human disease: Epstein–Barr virus (EBV), Hepatitis B/C/D virus (HBV/HCV/HDV), human herpesvirus 8 (HHV-8), human immunodeficiency virus (HIV), human T-cell leukemia virus type I (HTLV-I), human papillomavirus (HPV), herpes simplex virus-1/2 (HSV-1/2), and Varicella–Zoster virus (VZV). Current literature linking T-cell exhaustion with critical telomere lengths and immune senescence are discussed. The concept that enduring antigen stimulation leads to T-cell exhaustion that favors telomere attrition and a cell fate marked by enhanced T-cell senescence appears to be a common endpoint to chronic viral infections.
Collapse
Affiliation(s)
- Marcia Bellon
- Department of Pathology, Center for Viral Pathogenesis, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Christophe Nicot
- Department of Pathology, Center for Viral Pathogenesis, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
23
|
Henriksen EKK, Jørgensen KK, Kaveh F, Holm K, Hamm D, Olweus J, Melum E, Chung BK, Eide TJ, Lundin KEA, Boberg KM, Karlsen TH, Hirschfield GM, Liaskou E. Gut and liver T-cells of common clonal origin in primary sclerosing cholangitis-inflammatory bowel disease. J Hepatol 2017; 66:116-122. [PMID: 27647428 DOI: 10.1016/j.jhep.2016.09.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 09/01/2016] [Accepted: 09/04/2016] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS Recruitment of gut-derived memory T-cells to the liver is believed to drive hepatic inflammation in primary sclerosing cholangitis (PSC). However, whether gut-infiltrating and liver-infiltrating T-cells share T cell receptors (TCRs) and antigenic specificities is unknown. We used paired gut and liver samples from PSC patients with concurrent inflammatory bowel disease (PSC-IBD), and normal tissue samples from colon cancer controls, to assess potential T cell clonotype overlap between the two compartments. METHODS High-throughput sequencing of TCRβ repertoires was applied on matched colon, liver and blood samples from patients with PSC-IBD (n=10), and on paired tumor-adjacent normal gut and liver tissue samples from colon cancer patients (n=10). RESULTS An average of 9.7% (range: 4.7-19.9%) memory T cell clonotypes overlapped in paired PSC-IBD affected gut and liver samples, after excluding clonotypes present at similar frequencies in blood. Shared clonotypes constituted on average 16.0% (range: 8.7-32.6%) and 15.0% (range: 5.9-26.3%) of the liver and gut memory T-cells, respectively. A significantly higher overlap was observed between paired PSC-IBD affected samples (8.7%, p=0.0007) compared to paired normal gut and liver samples (3.6%), after downsampling to equal number of reads. CONCLUSION Memory T-cells of common clonal origin were detected in paired gut and liver samples of patients with PSC-IBD. Our data indicate that this is related to PSC-IBD pathogenesis, suggesting that memory T-cells driven by shared antigens are present in the gut and liver of PSC-IBD patients. Our findings support efforts to therapeutically target memory T cell recruitment in PSC-IBD. LAY SUMMARY Primary sclerosing cholangitis (PSC) is a devastating liver disease strongly associated with inflammatory bowel disease (IBD). The cause of PSC is unknown, but it has been suggested that the immune reactions in the gut and the liver are connected. Our data demonstrate for the first time that a proportion of the T-cells in the gut and the liver react to similar triggers, and that this proportion is particularly high in patients with PSC and IBD.
Collapse
Affiliation(s)
- Eva Kristine Klemsdal Henriksen
- Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway; Research Institute of Internal Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway; K.G. Jebsen Inflammation Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kristin Kaasen Jørgensen
- Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway; Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway
| | - Fatemeh Kaveh
- K.G. Jebsen Inflammation Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital Ullevål, Oslo, Norway
| | - Kristian Holm
- Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway; Research Institute of Internal Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway; K.G. Jebsen Inflammation Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - David Hamm
- Adaptive Biotechnologies Corp., Seattle, WA, USA
| | - Johanna Olweus
- K.G. Jebsen Inflammation Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway; K.G. Jebsen Center for Cancer Immunotherapy, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Espen Melum
- Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway; Research Institute of Internal Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway; K.G. Jebsen Inflammation Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Section of Gastroenterology, Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Brian K Chung
- Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Centre for Liver Research and NIHR Birmingham Biomedical Research Unit, Institute of Biomedical Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Tor J Eide
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Pathology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Knut E A Lundin
- Section of Gastroenterology, Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway; Centre for Immune Regulation, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kirsten Muri Boberg
- Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway; K.G. Jebsen Inflammation Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Section of Gastroenterology, Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Tom H Karlsen
- Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway; Research Institute of Internal Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway; K.G. Jebsen Inflammation Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Section of Gastroenterology, Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Gideon M Hirschfield
- Centre for Liver Research and NIHR Birmingham Biomedical Research Unit, Institute of Biomedical Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Evaggelia Liaskou
- Centre for Liver Research and NIHR Birmingham Biomedical Research Unit, Institute of Biomedical Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.
| |
Collapse
|
24
|
Maintenance of the EBV-specific CD8 + TCRαβ repertoire in immunosuppressed lung transplant recipients. Immunol Cell Biol 2016; 95:77-86. [PMID: 27507557 PMCID: PMC5214975 DOI: 10.1038/icb.2016.71] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Revised: 07/04/2016] [Accepted: 08/01/2016] [Indexed: 12/12/2022]
Abstract
Epstein-Barr virus (EBV) is one of the most common viruses in humans, capable of causing life-threatening infections and cancers in immunocompromised individuals. Although CD8+ T cells provide key protection against EBV, the persistence and dynamics of specific T-cell receptor (TCR) clones during immunosuppression in transplant patients is largely unknown. For the first time, we used a novel single-cell TCRαβ multiplex-nested reverse transcriptase PCR to dissect TCRαβ clonal diversity within GLCTLVAML (GLC)-specific CD8+ T cells in healthy individuals and immunocompromised lung transplant recipients. The GLC peptide presented by HLA-A*02:01 is one of the most immunogenic T-cell targets from the EBV proteome. We found that the GLC-specific TCRαβ repertoire was heavily biased toward TRAV5 and encompassed five classes of public TCRαβs, suggesting that these clonotypes are preferentially utilized following infection. We identified that a common TRAV5 was diversely paired with different TRAJ and TRBV/TRBJ genes, in both immunocompetent and immunocompromised individuals, with an average of 12 different TCRαβ clonotypes/donor. Moreover, pre-transplant GLC-specific TCRαβ repertoires were relatively stable over 1 year post transplant under immunosuppression in the absence or presence of EBV reactivation. In addition, we provide the first evidence of early GLC-specific CD8+ T cells at 87 days post transplant, which preceded clinical EBV detection at 242 days in an EBV-seronegative patient receiving a lung allograft from an EBV-seropositive donor. This was associated with a relatively stable TCRαβ repertoire after CD8+ T-cell expansion. Our findings provide insights into the composition and temporal dynamics of the EBV-specific TCRαβ repertoire in immunocompromised transplant patients and suggest that the early detection of EBV-specific T cells might be a predictor of ensuing EBV blood viremia.
Collapse
|
25
|
Merlo A, Dalla Santa S, Dolcetti R, Zanovello P, Rosato A. Reverse immunoediting: When immunity is edited by antigen. Immunol Lett 2016; 175:16-20. [PMID: 27131431 DOI: 10.1016/j.imlet.2016.04.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 04/22/2016] [Indexed: 12/30/2022]
Abstract
Immune selective pressure occurring during cancer immunoediting shapes tumor features revealed at clinical presentation. However, in the "Escape" phase, the tumor itself has the chance to influence the immunological response. Therefore, the capacity of the immune response to sculpt the tumor characteristics is only one side of the coin and even the opposite is likely true, i.e. that an antigen can shape the immune response in a sort of "reverse immunoediting". This reciprocal modeling probably occurs continuously, whenever the immune system encounters a tumor/foreign antigen, and can be operative in the pathogen/immune system interplay, thus possibly permeating the protective immunity as a whole. In line with this view, the characterization of a T cell response as well as the design of both active and passive immunotherapy strategies should also take into account all Ag features (type, load and presentation). Overall, we suggest that the "reverse immunoediting" hypothesis could help to dissect the complex interplay between antigens and the immune repertoire, and to improve the outcome of immunotherapeutic approaches, where T cell responses are manipulated and reprogrammed.
Collapse
Affiliation(s)
- Anna Merlo
- Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata, 64, 35128 Padua, Italy
| | - Silvia Dalla Santa
- Department of Surgery, Oncology and Gastroenterology, Via Gattamelata, 64, University of Padua, 35128 Padua, Italy
| | - Riccardo Dolcetti
- CRO-IRCCS, National Cancer Institute, Via F. Gallini, 2, 33081 Aviano, PN, Italy; University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland 4102, Australia
| | - Paola Zanovello
- Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata, 64, 35128 Padua, Italy; Department of Surgery, Oncology and Gastroenterology, Via Gattamelata, 64, University of Padua, 35128 Padua, Italy
| | - Antonio Rosato
- Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata, 64, 35128 Padua, Italy; Department of Surgery, Oncology and Gastroenterology, Via Gattamelata, 64, University of Padua, 35128 Padua, Italy.
| |
Collapse
|
26
|
Zhang XY, Collins GP, Soilleux E, Eyre TA. Acute EBV masquerading as peripheral T-cell lymphoma. BMJ Case Rep 2016; 2016:bcr-2015-213573. [PMID: 26838300 DOI: 10.1136/bcr-2015-213573] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Histological features of Epstein-Barr virus (EBV) can rarely mimic lymphoma. A 25-year-old presented with a spontaneous splenic rupture following a short illness. Histopathology assessment of the splenic and marrow tissue was highly suggestive of peripheral T-cell lymphoma not-otherwise-specified (PTCL-NOS). T-cell receptor (TCR) PCR clonality studies revealed a monoclonal T-cell population expressing for both TCRβ and γ, strongly suggestive of a T-cell clonal disease. EBV IgM was positive and IgG negative. EBV PCR was positive (7.02 ×10(4)/mL). Despite the strong suggestion of PTCL-NOS from histopathology and clonality studies, the decision was made to observe the patient and not start multiagent chemotherapy. The patient remained well, with no signs of PTCL and subsequently seroconverted to IgG+ EBV. We highlight the potential pitfall of acute EBV masquerading as PTCL and show the critical role of the multidisciplinary integration of histopathological, serology, molecular and clinical features to avoid misdiagnosis.
Collapse
Affiliation(s)
- Xiao-Yin Zhang
- Department of Haematology, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Graham P Collins
- Department of Haematology, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Elizabeth Soilleux
- Department of Cellular Pathology, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Toby Andrew Eyre
- Department of Haematology, Oxford University Hospitals NHS Trust, Oxford, UK
| |
Collapse
|
27
|
Cárdenas D, Vélez G, Orfao A, Herrera MV, Solano J, Olaya M, Uribe AM, Saavedra C, Duarte M, Rodríguez M, López M, Fiorentino S, Quijano S. Epstein-Barr virus-specific CD8(+) T lymphocytes from diffuse large B cell lymphoma patients are functionally impaired. Clin Exp Immunol 2015; 182:173-83. [PMID: 26174440 PMCID: PMC4608507 DOI: 10.1111/cei.12682] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2015] [Indexed: 12/13/2022] Open
Abstract
Epstein-Barr virus (EBV) is a persistent virus with oncogenic capacity that has been implicated in the development of aggressive B cell lymphomas, primarily in immunosuppressed individuals, although it can be present in immunocompetent individuals. Changes in the function and clonal diversity of T lymphocytes might be implied by viral persistence and lymphoma development. The aim of the present study was to evaluate the frequency, phenotype, function and clonotypical distribution of EBV-specific T cells after peripheral blood stimulation with a virus lysate in newly diagnosed patients with diffuse large B cell lymphoma (DLBCL) aged more than 50 years without prior histories of clinical immunosuppression compared with healthy controls. Our results showed impaired EBV-specific immune responses among DLBCL patients that were associated primarily with decreased numbers of central and effector memory CD8(+) T lymphocytes. In contrast to healthy controls, only a minority of the patients showed CD4(+)/tumour necrosis factor (TNF)-α(+) T cells expressing T cell receptor (TCR)-Vβ17 and CD8(+)/TNF-α(+) T cells with TCR-Vβ5·2, Vβ9 and Vβ18 in response to EBV. Notably, the production of TNF-α was undetectable among TCR-Vβ5·3(+), Vβ11(+), Vβ12(+), Vβ16(+) and Vβ23(+) CD8(+) T cells. In addition, we observed decreased numbers of CD4(+)/TNF-α(+) and CD8(+)/TNF-α(+), CD8(+)/interleukin (IL)-2(+) and CD8(+)/TNF-α(+)/IL-2(+) T lymphocytes in the absence of T cells capable of producing TNF-α, IL-2 and IFN-γ after EBV stimulation simultaneously. Moreover, DLBCL patients displayed higher IL-10 levels both under baseline conditions and after EBV stimulation. These findings were also observed in patients with positive EBV viral loads. Prospective studies including a large number of patients are needed to confirm these findings.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/virology
- Epstein-Barr Virus Infections/blood
- Epstein-Barr Virus Infections/immunology
- Epstein-Barr Virus Infections/virology
- Female
- Flow Cytometry
- Herpesvirus 4, Human/immunology
- Herpesvirus 4, Human/physiology
- Host-Pathogen Interactions/immunology
- Humans
- Interferon-gamma/immunology
- Interferon-gamma/metabolism
- Interleukin-10/immunology
- Interleukin-10/metabolism
- Interleukin-2/immunology
- Interleukin-2/metabolism
- Lymphocyte Count
- Lymphoma, Large B-Cell, Diffuse/blood
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/virology
- Male
- Middle Aged
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Tumor Necrosis Factor-alpha/immunology
- Tumor Necrosis Factor-alpha/metabolism
- Viral Load/immunology
Collapse
Affiliation(s)
- D Cárdenas
- Grupo De Inmunobiología Y Biología Celular Departamento De Microbiología Facultad De Ciencias Pontificia Universidad JaverianaBogotá, Colombia
| | - G Vélez
- Grupo De Inmunobiología Y Biología Celular Departamento De Microbiología Facultad De Ciencias Pontificia Universidad JaverianaBogotá, Colombia
| | - A Orfao
- Servicio General De Citometría Y Departamento De Medicina, Centro De Investigación Del Cáncer (Instituto De Biología Molecular Y Celular Del Cáncer and IBSAL; CSIC-USAL), Universidad De SalamancaSalamanca, España
| | - M V Herrera
- Servicio De Hematología Hospital Universitario San Ignacio-Centro De Oncología Javeriano
| | - J Solano
- Servicio De Hematología Hospital Universitario San Ignacio-Centro De Oncología Javeriano
| | - M Olaya
- Departamento de Patología, Pontificia Universidad Javeriana, Hospital Universitario San Ignacio
| | - A M Uribe
- Departamento de Patología, Pontificia Universidad Javeriana, Hospital Universitario San Ignacio
| | - C Saavedra
- Grupo De Patología Fundación Santa Fe De Bogotá
| | - M Duarte
- Servicio De Hematología Fundación Santa Fe De BogotáBogotá, Colombia
| | - M Rodríguez
- Servicio De Hematología Fundación Santa Fe De BogotáBogotá, Colombia
| | - M López
- Fundación Cardiovascular De ColombiaFloridablanca, Colombia
| | - S Fiorentino
- Grupo De Inmunobiología Y Biología Celular Departamento De Microbiología Facultad De Ciencias Pontificia Universidad JaverianaBogotá, Colombia
| | - S Quijano
- Grupo De Inmunobiología Y Biología Celular Departamento De Microbiología Facultad De Ciencias Pontificia Universidad JaverianaBogotá, Colombia
| |
Collapse
|
28
|
Moss DJ, Lutzky VP. EBV-Specific Immune Response: Early Research and Personal Reminiscences. Curr Top Microbiol Immunol 2015; 390:23-42. [PMID: 26424642 DOI: 10.1007/978-3-319-22822-8_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Early research on Epstein-Barr virus (EBV) developed from serological observations that were made soon after the discovery of the virus. Indeed, the definition of the humoral response to a variety of EBV proteins dominated the early literature and was instrumental in providing the key evidence for the association of the virus with infectious mononucleosis (IM), Burkitt's lymphoma (BL), and nasopharyngeal carcinoma (NPC). Each of these disease associations involved a distinct pattern of serological reactivity to the EBV membrane antigens (MA), early antigens (EA), and the EBV nuclear antigen (EBNA). When it became generally accepted that the marked lymphocytosis , which is a hallmark of acute IM, was dominated by T cells, considerable effort was directed toward untangling the specificities that might be associated with restricting the proliferation of newly infected B cells. Early evidence was divided between support for both EBV non-specific and/or HLA non-restricted components. However, all results needed to be reassessed in light of the observation that T cells died by apoptosis within hours of separation from fresh blood from acute IM patients. The observation that EBV-infected cultures from immune (but not non-immune) individuals began to die (termed regression) about 10 days post-seeding, provided the first evidence of a specific memory response which was apparently capable of controlling the small pool of latently infected B cells which all immune individuals possess. In this early era, CD8(+) T cells were thought to be the effector population responsible for this phenomenon, but later studies suggested a role for CD4(+) cells. This historical review includes reference to key early observations in regard to both the specific humoral and cellular responses to EBV infection from the time of the discovery of the virus until 1990. As well, we have included personal recollections in regard to the events surrounding the discovery of the memory T cell response since we believe they add a human dimension to a chapter focussed on early history.
Collapse
Affiliation(s)
- D J Moss
- QIMR Berghofer Medical Research Institute, Brisbane, Australia.
| | - V P Lutzky
- QIMR Berghofer Medical Research Institute, Brisbane, Australia.
| |
Collapse
|
29
|
Cárdenas Sierra D, Vélez Colmenares G, Orfao de Matos A, Fiorentino Gómez S, Quijano Gómez SM. Age-associated Epstein-Barr virus-specific T cell responses in seropositive healthy adults. Clin Exp Immunol 2014; 177:320-32. [PMID: 24666437 PMCID: PMC4089182 DOI: 10.1111/cei.12337] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2014] [Indexed: 12/13/2022] Open
Abstract
Epstein-Barr virus (EBV) is present in 95% of the world's adult population. The immune response participates in immune vigilance and persistent infection control, and this condition is maintained by both a good quality (functionality) and quantity of specific T cells throughout life. In the present study, we evaluated EBV-specific CD4(+) and CD8(+) T lymphocyte responses in seropositive healthy individuals younger and older than 50 years of age. The assessment comprised the frequency, phenotype, functionality and clonotypic distribution of T lymphocytes. We found that in both age groups a similar EBV-specific T cell response was found, with overlapping numbers of tumour necrosis factor (TNF)-α(+) T lymphocytes (CD4(+) and CD8(+)) within the memory and effector cell compartments, in addition to monofunctional and multi-functional T cells producing interleukin (IL)-2 and/or interferon (IFN)-γ. However, individuals aged more than 50 years showed significantly higher frequencies of IL-2-producing CD4(+) T lymphocytes in association with greater production of soluble IFN-γ, TNF-α and IL-6 than subjects younger than 50 years. A polyclonal T cell receptor (TCR)-variable beta region (Vβ) repertoire exists in both age groups under basal conditions and in response to EBV; the major TCR families found in TNF-α(+) /CD4(+) T lymphocytes were Vβ1, Vβ2, Vβ17 and Vβ22 in both age groups, and the major TCR family in TNF-α(+) /CD8(+) T cells was Vβ13·1 for individuals younger than 50 years and Vβ9 for individuals aged more than 50 years. Our findings suggest that the EBV-specific T cell response (using a polyclonal stimulation model) is distributed throughout several T cell differentiation compartments in an age-independent manner and includes both monofunctional and multi-functional T lymphocytes.
Collapse
Affiliation(s)
- D Cárdenas Sierra
- Grupo de Inmunobiología y Biología Celular, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | | | | | | |
Collapse
|
30
|
Iancu EM, Gannon PO, Laurent J, Gupta B, Romero P, Michielin O, Romano E, Speiser DE, Rufer N. Persistence of EBV antigen-specific CD8 T cell clonotypes during homeostatic immune reconstitution in cancer patients. PLoS One 2013; 8:e78686. [PMID: 24205294 PMCID: PMC3808305 DOI: 10.1371/journal.pone.0078686] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 09/15/2013] [Indexed: 11/19/2022] Open
Abstract
Persistent viruses are kept in check by specific lymphocytes. The clonal T cell receptor (TCR) repertoire against Epstein-Barr virus (EBV), once established following primary infection, exhibits a robust stability over time. However, the determinants contributing to this long-term persistence are still poorly characterized. Taking advantage of an in vivo clinical setting where lymphocyte homeostasis was transiently perturbed, we studied EBV antigen-specific CD8 T cells before and after non-myeloablative lympho-depleting chemotherapy of melanoma patients. Despite more advanced T cell differentiation, patients T cells showed clonal composition comparable to healthy individuals, sharing a preference for TRBV20 and TRBV29 gene segment usage and several co-dominant public TCR clonotypes. Moreover, our data revealed the presence of relatively few dominant EBV antigen-specific T cell clonotypes, which mostly persisted following transient lympho-depletion (TLD) and lymphocyte recovery, likely related to absence of EBV reactivation and de novo T cell priming in these patients. Interestingly, persisting clonotypes frequently co-expressed memory/homing-associated genes (CD27, IL7R, EOMES, CD62L/SELL and CCR5) supporting the notion that they are particularly important for long-lasting CD8 T cell responses. Nevertheless, the clonal composition of EBV-specific CD8 T cells was preserved over time with the presence of the same dominant clonotypes after non-myeloablative chemotherapy. The observed clonotype persistence demonstrates high robustness of CD8 T cell homeostasis and reconstitution.
Collapse
Affiliation(s)
- Emanuela M. Iancu
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Philippe O. Gannon
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Julien Laurent
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Bhawna Gupta
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Pedro Romero
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Lausanne, Switzerland
- Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Olivier Michielin
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Lausanne, Switzerland
- Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Emanuela Romano
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Daniel E. Speiser
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Lausanne, Switzerland
- Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Nathalie Rufer
- Department of Oncology, Lausanne University Hospital Center (CHUV) and University of Lausanne, Lausanne, Switzerland
- Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
31
|
Frigstad T, Løset GÅ, Sandlie I, Bogen B. A public T cell receptor recognized by a monoclonal antibody specific for the D-J junction of the β-chain. Scand J Immunol 2013; 78:345-51. [PMID: 23841814 DOI: 10.1111/sji.12098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 06/27/2013] [Indexed: 11/30/2022]
Abstract
It is becoming increasingly clear that T cell responses against many antigens are dominated by public α/β T cell receptors (TCRs) with restricted heterogeneity. Because expression of public TCRs may be related to resistance, or predisposition to diseases, it is relevant to measure their frequencies. Although staining with tetrameric peptide/major histocompatibility complex (pMHC) molecules gives information about specificity, it does not give information about the TCR composition of the individual T cells that stain. Moreover, next-generation sequencing of TCR does not yield information on pairing of α- and β-chains in single T cells. In an effort to overcome these limitations, we have here investigated the possibility of raising a monoclonal antibody (moAb) that recognizes a public TCR. As a model system, we have used T cells responding to the 91-101 CDR3 peptide of an Ig L-chain (λ2³¹⁵), presented by the MHC class II molecule I-E(d). The CD4⁺ T cell responses against this pMHC are dominated by a receptor composed of Vα3Jα1;Vβ6DβJβ1.1. Even the V(D)J junctions are to a large extent shared between T cell clones derived from different BALB/c mice. We here describe a murine moAb (AB10) of B10.D2 origin that recognizes this public TCR, while binding to peripheral T cells is negligible. Binding of the moAb is abrogated by introduction of two Gly residues in the D-J junction of the CDR3 of the β-chain. A model for the public TCR determinant is presented.
Collapse
Affiliation(s)
- T Frigstad
- Centre for Immune Regulation and Institute of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | | | | | | |
Collapse
|
32
|
Klarenbeek PL, Remmerswaal EBM, ten Berge IJM, Doorenspleet ME, van Schaik BDC, Esveldt REE, Koch SD, ten Brinke A, van Kampen AHC, Bemelman FJ, Tak PP, Baas F, de Vries N, van Lier RAW. Deep sequencing of antiviral T-cell responses to HCMV and EBV in humans reveals a stable repertoire that is maintained for many years. PLoS Pathog 2012; 8:e1002889. [PMID: 23028307 PMCID: PMC3460621 DOI: 10.1371/journal.ppat.1002889] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2012] [Accepted: 07/11/2012] [Indexed: 01/17/2023] Open
Abstract
CD8+ T-cell responses against latent viruses can cover considerable portions of the CD8+ T-cell compartment for many decades, yet their initiation and maintenance remains poorly characterized in humans. A key question is whether the clonal repertoire that is raised during the initial antiviral response can be maintained over these long periods. To investigate this we combined next-generation sequencing of the T-cell receptor repertoire with tetramer-sorting to identify, quantify and longitudinally follow virus-specific clones within the CD8+ T-cell compartment. Using this approach we studied primary infections of human cytomegalovirus (hCMV) and Epstein Barr virus (EBV) in renal transplant recipients. For both viruses we found that nearly all virus-specific CD8+ T-cell clones that appeared during the early phase of infection were maintained at high frequencies during the 5-year follow-up and hardly any new anti-viral clones appeared. Both in transplant recipients and in healthy carriers the clones specific for these latent viruses were highly dominant within the CD8+ T-cell receptor Vβ repertoire. These findings suggest that the initial antiviral response in humans is maintained in a stable fashion without signs of contraction or changes of the clonal repertoire. Several viruses have found ways to evade the human immune system and cause latent infections. Examples include HIV and herpes-viruses. Most humans carry these herpes-viruses. The human immune system mounts continuous responses against these viruses to prevent them from causing disease. If this balance is disturbed, these viruses can cause extensive pathology. We do not know how the immune response against these viruses evolves over time. Understanding this response might help to understand why the immune system does not clear these viruses and might help in preventive and therapeutic strategies. Here we used a new technology that allowed us to track virus specific immune cells (CD8+ T cells) over time in a quantitative manner. When we used this technology to study the evolution of latent responses against herpes-viruses (from infection until 5 years later) we found that immune responses were very rigid and did not evolve over time. Collectively our data shows that – for these herpes-viruses – the initial immune response is maintained despite the fact that this does not result in clearance of the virus. Therefore, if a virus survives the initial response, it will not be cleared in the future. This is an important consideration in understanding latent infection and for vaccination-design.
Collapse
Affiliation(s)
- P. L. Klarenbeek
- Department of Clinical Immunology & Rheumatology, Academic Medical Center, Amsterdam, the Netherlands
- Department of Genome Analysis, Academic Medical Center, Amsterdam, the Netherlands
- Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands
| | - E. B. M. Remmerswaal
- Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands
- Renal Transplant Unit, Academic Medical Center, Amsterdam, the Netherlands
| | - I. J. M. ten Berge
- Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands
- Renal Transplant Unit, Academic Medical Center, Amsterdam, the Netherlands
| | - M. E. Doorenspleet
- Department of Clinical Immunology & Rheumatology, Academic Medical Center, Amsterdam, the Netherlands
- Department of Genome Analysis, Academic Medical Center, Amsterdam, the Netherlands
- Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands
| | - B. D. C. van Schaik
- Bioinformatics Laboratory, Academic Medical Center, Amsterdam, the Netherlands
| | - R. E. E. Esveldt
- Department of Clinical Immunology & Rheumatology, Academic Medical Center, Amsterdam, the Netherlands
- Department of Genome Analysis, Academic Medical Center, Amsterdam, the Netherlands
- Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands
| | - S. D. Koch
- Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands
| | - A. ten Brinke
- Sanquin Research at CLB and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - A. H. C. van Kampen
- Bioinformatics Laboratory, Academic Medical Center, Amsterdam, the Netherlands
| | - F. J. Bemelman
- Renal Transplant Unit, Academic Medical Center, Amsterdam, the Netherlands
| | - P. P. Tak
- Department of Clinical Immunology & Rheumatology, Academic Medical Center, Amsterdam, the Netherlands
| | - F. Baas
- Department of Genome Analysis, Academic Medical Center, Amsterdam, the Netherlands
| | - N. de Vries
- Department of Clinical Immunology & Rheumatology, Academic Medical Center, Amsterdam, the Netherlands
| | - R. A. W. van Lier
- Department of Experimental Immunology, Academic Medical Center, Amsterdam, the Netherlands
- Sanquin Research at CLB and Landsteiner Laboratory, Amsterdam, the Netherlands
- * E-mail:
| |
Collapse
|
33
|
Torti N, Oxenius A. T cell memory in the context of persistent herpes viral infections. Viruses 2012; 4:1116-43. [PMID: 22852044 PMCID: PMC3407898 DOI: 10.3390/v4071116] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 07/18/2012] [Accepted: 07/19/2012] [Indexed: 12/16/2022] Open
Abstract
The generation of a functional memory T cell pool upon primary encounter with an infectious pathogen is, in combination with humoral immunity, an essential process to confer protective immunity against reencounters with the same pathogen. A prerequisite for the generation and maintenance of long-lived memory T cells is the clearance of antigen after infection, which is fulfilled upon resolution of acute viral infections. Memory T cells play also a fundamental role during persistent viral infections by contributing to relative control and immuosurveillance of active replication or viral reactivation, respectively. However, the dynamics, the phenotype, the mechanisms of maintenance and the functionality of memory T cells which develop upon acute/resolved infection as opposed to chronic/latent infection differ substantially. In this review we summarize current knowledge about memory CD8 T cell responses elicited during α-, β-, and γ-herpes viral infections with major emphasis on the induction, maintenance and function of virus-specific memory CD8 T cells during viral latency and we discuss how the peculiar features of these memory CD8 T cell responses are related to the biology of these persistently infecting viruses.
Collapse
Affiliation(s)
- Nicole Torti
- Institute of Microbiology, ETH Zurich, CH-8093 Zurich, Switzerland.
| | | |
Collapse
|
34
|
Connelley TK, MacHugh ND, Pelle R, Weir W, Morrison WI. Escape from CD8+ T cell response by natural variants of an immunodominant epitope from Theileria parva is predominantly due to loss of TCR recognition. THE JOURNAL OF IMMUNOLOGY 2011; 187:5910-20. [PMID: 22058411 DOI: 10.4049/jimmunol.1102009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Polymorphism of immunodominant CD8(+) T cell epitopes can facilitate escape from immune recognition of pathogens, leading to strain-specific immunity. In this study, we examined the TCR β-chain (TRB) diversity of the CD8(+) T cell responses of cattle against two immunodominant epitopes from Theileria parva (Tp1(214-224) and Tp2(49-59)) and investigated the role of TCR recognition and MHC binding in determining differential recognition of a series of natural variants of the highly polymorphic Tp2(49-59) epitope by CD8(+) T cell clones of defined TRB genotype. Our results show that both Tp1(214-224) and Tp2(49-59) elicited CD8(+) T cell responses using diverse TRB repertoires that showed a high level of stability following repeated pathogenic challenge over a 3-y period. Analysis of single-alanine substituted versions of the Tp2(49-59) peptide demonstrated that Tp2(49-59)-specific clonotypes had a broad range of fine specificities for the epitope. Despite this diversity, all natural variants exhibited partial or total escape from immune recognition, which was predominantly due to abrogation of TCR recognition, with mutation resulting in loss of the lysine residue at P8, playing a particularly dominant role in escape. The levels of heterozygosity in individual Tp2(49-59) residues correlated closely with loss of immune recognition, suggesting that immune selection has contributed to epitope polymorphism.
Collapse
Affiliation(s)
- Timothy K Connelley
- The Roslin Institute and The Royal Dick School of Veterinary Studies, University of Edinburgh, Midlothian EH25 9RG, Scotland, United Kingdom.
| | | | | | | | | |
Collapse
|
35
|
Terajima M, Ennis FA. T cells and pathogenesis of hantavirus cardiopulmonary syndrome and hemorrhagic fever with renal syndrome. Viruses 2011; 3:1059-73. [PMID: 21994770 PMCID: PMC3185782 DOI: 10.3390/v3071059] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Revised: 06/30/2011] [Accepted: 07/01/2011] [Indexed: 12/17/2022] Open
Abstract
We previously hypothesized that increased capillary permeability observed in both hantavirus cardiopulmonary syndrome (HCPS) and hemorrhagic fever with renal syndrome (HFRS) may be caused by hantavirus-specific cytotoxic T cells attacking endothelial cells presenting viral antigens on their surface based on clinical observations and in vitro experiments. In HCPS, hantavirus-specific T cell responses positively correlated with disease severity. In HFRS, in one report, contrary to HCPS, T cell responses negatively correlated with disease severity, but in another report the number of regulatory T cells, which are thought to suppress T cell responses, negatively correlated with disease severity. In rat experiments, in which hantavirus causes persistent infection, depletion of regulatory T cells helped infected rats clear virus without inducing immunopathology. These seemingly contradictory findings may suggest delicate balance in T cell responses between protection and immunopathogenesis. Both too strong and too weak T cell responses may lead to severe disease. It is important to clarify the role of T cells in these diseases for better treatment (whether to suppress T cell functions) and protection (vaccine design) which may need to take into account viral factors and the influence of HLA on T cell responses.
Collapse
Affiliation(s)
- Masanori Terajima
- Center for Infectious Disease and Vaccine Research, Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA; E-Mail:
| | - Francis A. Ennis
- Center for Infectious Disease and Vaccine Research, Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA; E-Mail:
| |
Collapse
|
36
|
Miles JJ, Douek DC, Price DA. Bias in the αβ T-cell repertoire: implications for disease pathogenesis and vaccination. Immunol Cell Biol 2011; 89:375-87. [PMID: 21301479 DOI: 10.1038/icb.2010.139] [Citation(s) in RCA: 167] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The naïve T-cell repertoire is vast, containing millions of unique T-cell receptor (TCR) structures. Faced with such diversity, the mobilization of TCR structures from this enormous pool was once thought to be a stochastic, even chaotic, process. However, steady and systematic dissection over the last 20 years has revealed that this is not the case. Instead, the TCR repertoire deployed against individual antigens is routinely ordered and biased. Often, identical and near-identical TCR repertoires can be observed across different individuals, suggesting that the system encompasses an element of predictability. This review provides a catalog of αβ TCR bias by disease and by species, and discusses the mechanisms that govern this inherent and widespread phenomenon.
Collapse
Affiliation(s)
- John J Miles
- T Cell Modulation Laboratory, Department of Infection, Immunity and Biochemistry, Cardiff University School of Medicine, Cardiff, UK.
| | | | | |
Collapse
|
37
|
Burrows SR, Moss DJ, Khanna R. Understanding human T-cell-mediated immunoregulation through herpesviruses. Immunol Cell Biol 2011; 89:352-8. [PMID: 21301481 DOI: 10.1038/icb.2010.136] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Human herpesviruses have coevolved with humans over millions of years, and adaptation of latent infection within the cells of the immune system is a unique characteristic of many of these viruses. Following primary infection, these herpesviruses establish an asymptomatic-persistent infection in healthy individuals that is strictly controlled by virus-specific CD8(+) and CD4(+) T cells. Here, we provide a brief overview of how the human immune system interacts with these latent viruses and regulates the lifelong host-virus relationship in healthy virus carriers. Extensive studies on T-cell-mediated immune regulation over the last decade has allowed researchers to successfully translate these findings into the clinical setting to treat various herpesvirus-associated diseases in transplant patients and individuals with virus-associated malignancies. It is highly likely that these newly emerging T-cell-based therapeutic and diagnostic technologies will revolutionize the clinical management of patients with herpesvirus-associated diseases.
Collapse
Affiliation(s)
- Scott R Burrows
- Australian Centre for Vaccine Development, Queensland Institute of Medical Research, Brisbane, Queensland, Australia.
| | | | | |
Collapse
|
38
|
Miles JJ, Thammanichanond D, Moneer S, Nivarthi UK, Kjer-Nielsen L, Tracy SL, Aitken CK, Brennan RM, Zeng W, Marquart L, Jackson D, Burrows SR, Bowden DS, Torresi J, Hellard M, Rossjohn J, McCluskey J, Bharadwaj M. Antigen-driven patterns of TCR bias are shared across diverse outcomes of human hepatitis C virus infection. THE JOURNAL OF IMMUNOLOGY 2010; 186:901-12. [PMID: 21160049 DOI: 10.4049/jimmunol.1003167] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hepatitis C virus (HCV) infection causes significant morbidity and mortality worldwide. T cells play a central role in HCV clearance; however, there is currently little understanding of whether the disease outcome in HCV infection is influenced by the choice of TCR repertoire. TCR repertoires used against two immunodominant HCV determinants--the highly polymorphic, HLA-B*0801 restricted (1395)HSKKKCDEL(1403) (HSK) and the comparatively conserved, HLA-A*0101-restricted, (1435)ATDALMTGY(1443) (ATD)--were analyzed in clearly defined cohorts of HLA-matched, HCV-infected individuals with persistent infection and HCV clearance. In comparison with ATD, TCR repertoire selected against HSK was more narrowly focused, supporting reports of mutational escape in this epitope, in persistent HCV infection. Notwithstanding the Ag-driven divergence, T cell repertoire selection against either Ag was comparable in subjects with diverse disease outcomes. Biased T cell repertoires were observed early in infection and were evident not only in persistently infected individuals but also in subjects with HCV clearance, suggesting that these are not exclusively characteristic of viral persistence. Comprehensive clonal analysis of Ag-specific T cells revealed widespread use of public TCRs displaying a high degree of predictability in TRBV/TRBJ gene usage, CDR3 length, and amino acid composition. These public TCRs were observed against both ATD and HSK and were shared across diverse disease outcomes. Collectively, these observations indicate that repertoire diversity rather than particular Vβ segments are better associated with HCV persistence/clearance in humans. Notably, many of the anti-HCV TCRs switched TRBV and TRBJ genes around a conserved, N nucleotide-encoded CDR3 core, revealing TCR sequence mosaicism as a potential host mechanism to combat this highly variant virus.
Collapse
Affiliation(s)
- John J Miles
- Queensland Institute of Medical Research, Queensland 4029, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Miles JJ, Bulek AM, Cole DK, Gostick E, Schauenburg AJA, Dolton G, Venturi V, Davenport MP, Tan MP, Burrows SR, Wooldridge L, Price DA, Rizkallah PJ, Sewell AK. Genetic and structural basis for selection of a ubiquitous T cell receptor deployed in Epstein-Barr virus infection. PLoS Pathog 2010; 6:e1001198. [PMID: 21124993 PMCID: PMC2987824 DOI: 10.1371/journal.ppat.1001198] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Accepted: 10/15/2010] [Indexed: 12/02/2022] Open
Abstract
Despite the ∼10(18) αβ T cell receptor (TCR) structures that can be randomly manufactured by the human thymus, some surface more frequently than others. The pinnacles of this distortion are public TCRs, which exhibit amino acid-identical structures across different individuals. Public TCRs are thought to result from both recombinatorial bias and antigen-driven selection, but the mechanisms that underlie inter-individual TCR sharing are still largely theoretical. To examine this phenomenon at the atomic level, we solved the co-complex structure of one of the most widespread and numerically frequent public TCRs in the human population. The archetypal AS01 public TCR recognizes an immunodominant BMLF1 peptide, derived from the ubiquitous Epstein-Barr virus, bound to HLA-A*0201. The AS01 TCR was observed to dock in a diagonal fashion, grasping the solvent exposed peptide crest with two sets of complementarity-determining region (CDR) loops, and was fastened to the peptide and HLA-A*0201 platform with residue sets found only within TCR genes biased in the public response. Computer simulations of a random V(D)J recombination process demonstrated that both TCRα and TCRβ amino acid sequences could be manufactured easily, thereby explaining the prevalence of this receptor across different individuals. Interestingly, the AS01 TCR was encoded largely by germline DNA, indicating that the TCR loci already comprise gene segments that specifically recognize this ancient pathogen. Such pattern recognition receptor-like traits within the αβ TCR system further blur the boundaries between the adaptive and innate immune systems.
Collapse
MESH Headings
- Amino Acid Sequence
- Antigens, Viral/immunology
- CD8-Positive T-Lymphocytes
- Computer Simulation
- Crystallization
- Crystallography, X-Ray
- Cytotoxicity, Immunologic
- HLA-A Antigens/immunology
- HLA-A2 Antigen
- Herpesviridae Infections/immunology
- Herpesviridae Infections/metabolism
- Herpesviridae Infections/virology
- Herpesvirus 4, Human/immunology
- Humans
- Immune Tolerance
- Molecular Sequence Data
- Protein Conformation
- Receptors, Antigen, T-Cell, alpha-beta/chemistry
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Recombination, Genetic
- Sequence Homology, Amino Acid
- Surface Plasmon Resonance
Collapse
Affiliation(s)
- John J Miles
- Department of Infection, Cardiff University School of Medicine, Heath Park, Cardiff, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Clute SC, Naumov YN, Watkin LB, Aslan N, Sullivan JL, Thorley-Lawson DA, Luzuriaga K, Welsh RM, Puzone R, Celada F, Selin LK. Broad cross-reactive TCR repertoires recognizing dissimilar Epstein-Barr and influenza A virus epitopes. THE JOURNAL OF IMMUNOLOGY 2010; 185:6753-64. [PMID: 21048112 DOI: 10.4049/jimmunol.1000812] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Memory T cells cross-reactive with epitopes encoded by related or even unrelated viruses may alter the immune response and pathogenesis of infection by a process known as heterologous immunity. Because a challenge virus epitope may react with only a subset of the T cell repertoire in a cross-reactive epitope-specific memory pool, the vigorous cross-reactive response may be narrowly focused, or oligoclonal. We show in this article, by examining human T cell cross-reactivity between the HLA-A2-restricted influenza A virus-encoded M1(58-66) epitope (GILGFVFTL) and the dissimilar Epstein-Barr virus-encoded BMLF1(280-288) epitope (GLCTLVAML), that, under some conditions, heterologous immunity can lead to a significant broadening, rather than a narrowing, of the TCR repertoire. We suggest that dissimilar cross-reactive epitopes might generate a broad, rather than a narrow, T cell repertoire if there is a lack of dominant high-affinity clones; this hypothesis is supported by computer simulation.
Collapse
Affiliation(s)
- Shalyn C Clute
- Department of Pathology and Program in Immunology and Virology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Sircar P, Furr KL, Dorosh LA, Letvin NL. Clonal Repertoires of Virus-Specific CD8+T Lymphocytes Are Shared in Mucosal and Systemic Compartments during Chronic Simian Immunodeficiency Virus Infection in Rhesus Monkeys. THE JOURNAL OF IMMUNOLOGY 2010; 185:2191-9. [DOI: 10.4049/jimmunol.1001340] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
42
|
Jaquiéry E, Jilek S, Schluep M, Meylan P, Lysandropoulos A, Pantaleo G, Du Pasquier RA. Intrathecal immune responses to EBV in early MS. Eur J Immunol 2010; 40:878-87. [PMID: 20017197 DOI: 10.1002/eji.200939761] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
EBV has been consistently associated with MS, but its signature in the CNS has rarely been examined. In this study, we assessed EBV-specific humoral and cellular immune responses in the cerebrospinal fluid (CSF) of patients with early MS, other inflammatory neurological diseases (OIND) and non-inflammatory neurological diseases (NIND). The neurotropic herpesvirus CMV served as a control. Virus-specific humoral immune responses were assessed in 123 consecutive patients and the intrathecal recruitment of virus-specific antibodies was expressed as antibody indexes. Cellular immune responses tested in the blood of 55/123 patients were positive in 46/55. The CD8(+) CTL responses of these 46 patients were assessed in the blood and CSF using a CFSE-based CTL assay. We found that viral capsid antigen and EBV-encoded nuclear antigen-1, but not CMV IgG antibody indexes, were increased in early MS as compared with OIND and NIND patients. There was also intrathecal enrichment in EBV-, but not CMV-specific, CD8(+) CTL in early MS patients. By contrast, OIND and NIND patients did not recruit EBV- nor CMV-specific CD8(+) CTL in the CSF. Our data, showing a high EBV-, but not CMV-specific intrathecal immune response, strengthen the association between EBV and MS, in particular at the onset of the disease.
Collapse
Affiliation(s)
- Emilie Jaquiéry
- Service of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
43
|
Henson SM, Akbar AN. Memory T-Cell Homeostasis and Senescence during Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 684:189-97. [DOI: 10.1007/978-1-4419-6451-9_15] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
44
|
Isakov D, Dzutsev A, Belyakov IM, Berzofsky JA. Non-equilibrium and differential function between intraepithelial and lamina propria virus-specific TCRalphabeta(+) CD8alphabeta(+) T cells in the small intestinal mucosa. Mucosal Immunol 2009; 2:450-61. [PMID: 19571797 PMCID: PMC3224999 DOI: 10.1038/mi.2009.95] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The gastrointestinal mucosa regularly encounters commensal and pathogenic microbiota. Gut mucosal lymphocytes consist of two phenotypically different populations residing in the intestinal intraepithelial (IEL) compartment and lamina propria (LP). Little is known about compositional and functional differences of antigen-specific T cells from these mucosal compartments after mucosal infection, or the degree of trafficking between them. We here studied the B8R(20-27)-specific CD8 T-cell response in LP and IEL compartments after intrarectal immunization with modified vaccinia virus Ankara (MVA). CD8(+) T cells in the IEL compartment had much lower avidity than in the LP or spleen during acute and memory phases. Surprisingly, the TCR Vbeta-chain distribution of antigen-specific T cells and the length of the CDR3 region of the dominant Vbeta genes showed substantial dissimilarities between IEL and LP antigen-specific CD8alphabeta T cells in individual mice, increasing with time. We show functional and compositional differences between these mucosal compartments during the effector and memory phases of the immune response, indicating limited crosstalk and microenvironmental differences between the IEL, LP, and spleen. The restricted migration of cells from each of these mucosal compartments could partly account for a founder effect we observed in the IEL TCRalphabeta CD8alphabeta epitope-specific repertoire that might impact protective efficacy.
Collapse
Affiliation(s)
- D Isakov
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - A Dzutsev
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - IM Belyakov
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, USA, Midwest Research Institute, Frederick, Maryland, USA
| | - JA Berzofsky
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
45
|
Iancu EM, Corthesy P, Baumgaertner P, Devevre E, Voelter V, Romero P, Speiser DE, Rufer N. Clonotype selection and composition of human CD8 T cells specific for persistent herpes viruses varies with differentiation but is stable over time. THE JOURNAL OF IMMUNOLOGY 2009; 183:319-31. [PMID: 19542443 DOI: 10.4049/jimmunol.0803647] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Protection from reactivation of persistent herpes virus infection is mediated by Ag-specific CD8 T cell responses, which are highly regulated by still poorly understood mechanisms. In this study, we analyzed differentiation and clonotypic dynamics of EBV- and CMV-specific T cells from healthy adults. Although these T lymphocytes included all subsets, from early-differentiated (EM/CD28(pos)) to late-differentiated (EMRA/CD28(neg)) stages, they varied in the sizes/proportions of these subsets. In-depth clonal composition analyses revealed TCR repertoires, which were highly restricted for CMV- and relatively diverse for EBV-specific cells. Virtually all virus-specific clonotypes identified in the EMRA/CD28(neg) subset were also found within the pool of less differentiated "memory" cells. However, striking differences in the patterns of dominance were observed among these subsets, because some clonotypes were selected with differentiation while others were not. Late-differentiated CMV-specific clonotypes were mostly characterized by TCR with lower dependency on CD8 coreceptor interaction. Yet all clonotypes displayed similar functional avidities, suggesting a compensatory role of CD8 in the clonotypes of lower TCR avidity. Importantly, clonotype selection and composition of each virus-specific subset upon differentiation was highly preserved over time, with the presence of the same dominant clonotypes at specific differentiation stages within a period of 4 years. Remarkably, clonotypic distribution was stable not only in late-differentiated but also in less-differentiated T cell subsets. Thus, T cell clonotypes segregate with differentiation, but the clonal composition once established is kept constant for at least several years. These findings reveal novel features of the highly sophisticated control of steady state protective T cell activity in healthy adults.
Collapse
|
46
|
Upregulation of interleukin 7 receptor alpha and programmed death 1 marks an epitope-specific CD8+ T-cell response that disappears following primary Epstein-Barr virus infection. J Virol 2009; 83:9068-78. [PMID: 19605492 DOI: 10.1128/jvi.00141-09] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In immunocompetent individuals, the stability of the herpesvirus-host balance limits opportunities to study the disappearance of a virus-specific CD8(+) T-cell response. However, we noticed that in HLA-A 0201-positive infectious mononucleosis (IM) patients undergoing primary Epstein-Barr virus (EBV) infection, the initial CD8 response targets three EBV lytic antigen-derived epitopes, YVLDHLIVV (YVL), GLCTLVAML (GLC), and TLDYKPLSV (TLD), but only the YVL and GLC reactivities persist long-term; the TLD response disappears within 10 to 27 months. While present, TLD-specific cells remained largely indistinguishable from YVL and GLC reactivities in many phenotypic and functional respects but showed unique temporal changes in two markers of T-cell fate, interleukin 7 receptor alpha (IL-7Ralpha; CD127) and programmed death 1 (PD-1). Thus, following the antigen-driven downregulation of IL-7Ralpha seen on all populations in acute IM, in every case, the TLD-specific population recovered expression unusually quickly post-IM. As well, in four of six patients studied, TLD-specific cells showed very strong PD-1 upregulation in the last blood sample obtained before the cells' disappearance. Our data suggest that the disappearance of this individual epitope reactivity from an otherwise stable EBV-specific response (i) reflects a selective loss of cognate antigen restimulation (rather than of IL-7-dependent signals) and (ii) is immediately preceded, and perhaps mediated, by PD-1 upregulation to unprecedented levels.
Collapse
|
47
|
Co MDT, Kilpatrick ED, Rothman AL. Dynamics of the CD8 T-cell response following yellow fever virus 17D immunization. Immunology 2009; 128:e718-27. [PMID: 19740333 DOI: 10.1111/j.1365-2567.2009.03070.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Management of yellow fever is focused on the prevention of illness by the use of the yellow fever virus (YFV) 17D vaccine. The role of neutralizing antibodies in protection is generally accepted with YFV-specific T cells likely contributing to the control of viral replication. We studied CD8(+) T-cell responses to four defined human leucocyte antigen-B35-restricted epitopes in YFV vaccine recipients as a model of the kinetics of cytotoxic T-lymphocyte responses to an acute human viral infection. Multiple features of these epitope-specific responses were analysed after vaccination including magnitude, cytokine production, phenotype and T-cell receptor repertoire. Peak peptide-specific interferon-gamma (IFN-gamma) responses of almost 1% of CD8(+) T cells were seen as early as 2 weeks post-vaccination; however, dominant responses varied between donors. Peptide-specific responses were still detectable at 54 months post-vaccination. Tetramer-positive cells, at high frequencies, were detected as early as 7-9 days, before detectable IFN-gamma-producing cells, suggesting a defect in the functional capacity of some antigen-specific cells early post-vaccination. The predominant memory phenotype of the tetramer-positive population was a differentiated effector (CD45RA(+) CCR7(-) CD62L(-)) phenotype. The T-cell receptor Vbeta analysis revealed a diverse oligoclonal repertoire in tetramer-positive T-cell populations in two individuals. These characteristics of the YFV-specific T-cell response could contribute to vaccine effectiveness.
Collapse
Affiliation(s)
- Mary Dawn T Co
- Center for Infectious Disease and Vaccine Research, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | | | | |
Collapse
|
48
|
Venturi V, Chin HY, Asher TE, Ladell K, Scheinberg P, Bornstein E, van Bockel D, Kelleher AD, Douek DC, Price DA, Davenport MP. TCR beta-chain sharing in human CD8+ T cell responses to cytomegalovirus and EBV. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:7853-62. [PMID: 19017975 DOI: 10.4049/jimmunol.181.11.7853] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The CD8(+) TCR repertoires specific for many immunogenic epitopes of CMV and EBV are dominated by a few TCR clonotypes and involve public TCRs that are shared between many MHC-matched individuals. In previous studies, we demonstrated that the observed sharing of epitope-specific TCRbeta chains between individuals is strongly associated with TCRbeta production frequency, and that a process of convergent recombination facilitates the more efficient production of some TCRbeta sequences. In this study, we analyzed a total of 2836 TCRbeta sequences from 23 CMV-infected and 10 EBV-infected individuals to investigate the factors that influence the sharing of TCRbeta sequences in the CD8(+) T cell responses to two immunodominant HLA-A*0201-restricted epitopes from these viruses. The most shared TCRbeta amino acid sequences were found to have two features that indicate efficient TCRbeta production, as follows: 1) they required fewer nucleotide additions, and 2) they were encoded by a greater variety of nucleotide sequences. We used simulations of random V(D)J recombination to demonstrate that the in silico TCRbeta production frequency was predictive of the extent to which both TCRbeta nucleotide and amino acid sequences were shared in vivo. These results suggest that TCRbeta production frequency plays an important role in the interindividual sharing of TCRbeta sequences within CD8(+) T cell responses specific for CMV and EBV.
Collapse
Affiliation(s)
- Vanessa Venturi
- Complex Systems in Biology Group, Centre for Vascular Research, University of New South Wales, Kensington, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
CD8+ T cell immunity to Epstein-Barr virus and Kaposi's sarcoma-associated herpes virus. Semin Cancer Biol 2008; 18:416-22. [PMID: 19007888 DOI: 10.1016/j.semcancer.2008.10.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2008] [Accepted: 10/16/2008] [Indexed: 12/27/2022]
Abstract
Gammaherpesviruses are agents which have evolved to persist within the lymphoid system and many have oncogenic potential; studying gammaherpesvirus infections therefore has the potential to reveal much about the workings of the immune system and the control over viral oncogenesis. The lymphocryptovirus Epstein-Barr virus (EBV) and the rhadinovirus Kaposi's sarcoma-associated herpesvirus (KSHV, also known as human herpesvirus 8) are the two human gammaherpesviruses. Analysis of the T cell response to EBV has guided understanding of immunity to infection and disease caused by this virus, as well as directed the development of vaccination and therapeutic interventions in EBV-associated disease. Less is known about the T cell response to KSHV and its exact role in controlling virus infection and disease. Here we discuss the CD8+ T cell response to these two gammaherpesviruses.
Collapse
|
50
|
Venturi V, Chin HY, Price DA, Douek DC, Davenport MP. The role of production frequency in the sharing of simian immunodeficiency virus-specific CD8+ TCRs between macaques. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:2597-609. [PMID: 18684950 DOI: 10.4049/jimmunol.181.4.2597] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
In some epitope-specific responses, T cells bearing identical TCRs occur in many MHC-matched individuals. The sharing of public TCRs is unexpected, given the enormous potential diversity of the TCR repertoire. We have previously studied the sharing of TCR beta-chains in the CD8(+) T cell responses to two influenza epitopes in mice. Analysis of these TCRbeta repertoires suggests that, even with unbiased V(D)J recombination mechanisms, some TCRbetas can be produced more frequently than others, by a process of convergent recombination. The TCRbeta production frequency was shown to be a good predictor of the observed sharing of epitope-specific TCRbetas between mice. However, this study was limited to immune responses in an inbred population. In this study, we investigated TCRbeta sharing in CD8(+) T cell responses specific for the immunodominant Mamu-A*01-restricted Tat-SL8/TL8 and Gag-CM9 epitopes of SIV in rhesus macaques. Multiple data sets were used, comprising a total of approximately 6000 TCRbetas sampled from 20 macaques. We observed a spectrum in the number of macaques sharing epitope-specific TCRbetas in this outbred population. This spectrum of TCRbeta sharing was negatively correlated with the minimum number of nucleotide additions required to produce the sequences and strongly positively correlated with the number of observed nucleotide sequences encoding the amino acid sequences. We also found that TCRbeta sharing was correlated with the number of times, and the variety of different ways, the sequences were produced in silico via random gene recombination. Thus, convergent recombination is a major determinant of the extent of TCRbeta sharing.
Collapse
MESH Headings
- Amino Acid Motifs
- Animals
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/virology
- Epitopes, T-Lymphocyte/biosynthesis
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Gene Rearrangement, beta-Chain T-Cell Antigen Receptor
- Histocompatibility Antigens Class I/biosynthesis
- Histocompatibility Antigens Class I/genetics
- Histocompatibility Antigens Class I/immunology
- Macaca mulatta
- Peptide Fragments/biosynthesis
- Peptide Fragments/genetics
- Peptide Fragments/immunology
- Receptors, Antigen, T-Cell, alpha-beta/biosynthesis
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Sequence Alignment
- Simian Immunodeficiency Virus/immunology
Collapse
Affiliation(s)
- Vanessa Venturi
- Complex Systems in Biology Group, Centre for Vascular Research, University of New South Wales, Kensington, Australia
| | | | | | | | | |
Collapse
|