51
|
Sendi P, Hirzel C, Pfister S, Ackermann-Gäumann R, Grandgirard D, Hewer E, Nirkko AC. Fatal Outcome of European Tick-borne Encephalitis after Vaccine Failure. Front Neurol 2017; 8:119. [PMID: 28421031 PMCID: PMC5377060 DOI: 10.3389/fneur.2017.00119] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 03/15/2017] [Indexed: 12/30/2022] Open
Abstract
Tick-borne encephalitis is a viral disease affecting the central nervous system. It is endemic in Switzerland with 200–250 notified cases annually. Active immunization is effective for persons in all age groups. Vaccine failure is rare, in particular after a completed vaccination course. Here, we describe the case of 67-year-old man with a fatal outcome despite vaccination. The diagnosis was confirmed by extensive postmortem analyses. The diagnostic challenges of vaccine failure in tick-borne encephalitis and the dynamics of the immune response in vaccination breakthrough are discussed.
Collapse
Affiliation(s)
- Parham Sendi
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland.,Bern University Hospital, Department of Infectious Diseases, Bern, Switzerland
| | - Cédric Hirzel
- Bern University Hospital, Department of Infectious Diseases, Bern, Switzerland
| | - Stefan Pfister
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Rahel Ackermann-Gäumann
- Spiez Laboratory, Swiss National Reference Centre for Tick-transmitted Diseases, Federal Office for Civil Protection, Spiez, Switzerland
| | - Denis Grandgirard
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Ekkehard Hewer
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Arto C Nirkko
- Department of Neurology, Schlaf-Wach-Epilepsie-Zentrum (SWEZ), University of Bern, Bern, Switzerland
| |
Collapse
|
52
|
He R, Yang X, Liu C, Chen X, Wang L, Xiao M, Ye J, Wu Y, Ye L. Efficient control of chronic LCMV infection by a CD4 T cell epitope-based heterologous prime-boost vaccination in a murine model. Cell Mol Immunol 2017; 15:815-826. [PMID: 28287115 DOI: 10.1038/cmi.2017.3] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 12/08/2016] [Accepted: 12/08/2016] [Indexed: 02/07/2023] Open
Abstract
CD4+ T cells are essential for sustaining CD8+ T cell responses during a chronic infection. The adoptive transfer of virus-specific CD4+ T cells has been shown to efficiently rescue exhausted CD8+ T cells. However, the question of whether endogenous virus-specific CD4+ T cell responses can be enhanced by certain vaccination strategies and subsequently reinvigorate exhausted CD8+ T cells remains unexplored. In this study, we developed a CD4+ T cell epitope-based heterologous prime-boost immunization strategy and examined the efficacy of this strategy using a mouse model of chronic lymphocytic choriomeningitis virus (LCMV) infection. We primed chronically LCMV-infected mice with a Listeria monocytogenes vector that expressed the LCMV glycoprotein-specific I-Ab-restricted CD4+ T cell epitope GP61-80 (LM-GP61) and subsequently boosted the primed mice with an influenza virus A (PR8 strain) vector that expressed the same CD4+ T cell epitope (IAV-GP61). This heterologous prime-boost vaccination strategy elicited strong anti-viral CD4+ T cell responses, which further improved both the quantity and quality of the virus-specific CD8+ T cells and led to better control of the viral loads. The combination of this strategy and the blockade of the programmed cell death-1 (PD-1) inhibitory pathway further enhanced the anti-viral CD8+ T cell responses and viral clearance. Thus, a heterologous prime-boost immunization that selectively induces virus-specific CD4+ T cell responses in conjunction with blockade of the inhibitory pathway may represent a promising therapeutic approach to treating patients with chronic viral infections.
Collapse
Affiliation(s)
- Ran He
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, 510515, Guangzhou, China.,Institute of Immunology, Medical School, Third Military Medical University, 400038, Chongqing, China
| | - Xinxin Yang
- Institute of Immunology, Medical School, Third Military Medical University, 400038, Chongqing, China
| | - Cheng Liu
- Institute of Immunology, Medical School, Third Military Medical University, 400038, Chongqing, China
| | - Xiangyu Chen
- Institute of Immunology, Medical School, Third Military Medical University, 400038, Chongqing, China
| | - Lin Wang
- Institute of Immunology, Medical School, Third Military Medical University, 400038, Chongqing, China
| | - Minglu Xiao
- Institute of Immunology, Medical School, Third Military Medical University, 400038, Chongqing, China
| | - Jianqiang Ye
- Ministry of Education Key Laboratory for Avian Preventive Medicine, College of Veterinary Medicine, Yangzhou University, 225009, Yangzhou, China, Jiangsu.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, 225009, Yangzhou, China, Jiangsu
| | - Yuzhang Wu
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, 510515, Guangzhou, China.,Institute of Immunology, Medical School, Third Military Medical University, 400038, Chongqing, China
| | - Lilin Ye
- Institute of Immunology, Medical School, Third Military Medical University, 400038, Chongqing, China.
| |
Collapse
|
53
|
Pfeiler S, Khandagale AB, Magenau A, Nichols M, Heijnen HFG, Rinninger F, Ziegler T, Seveau S, Schubert S, Zahler S, Verschoor A, Latz E, Massberg S, Gaus K, Engelmann B. Distinct surveillance pathway for immunopathology during acute infection via autophagy and SR-BI. Sci Rep 2016; 6:34440. [PMID: 27694929 PMCID: PMC5046072 DOI: 10.1038/srep34440] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 09/13/2016] [Indexed: 12/21/2022] Open
Abstract
The mechanisms protecting from immunopathology during acute bacterial infections are incompletely known. We found that in response to apoptotic immune cells and live or dead Listeria monocytogenes scavenger receptor BI (SR-BI), an anti-atherogenic lipid exchange mediator, activated internalization mechanisms with characteristics of macropinocytosis and, assisted by Golgi fragmentation, initiated autophagic responses. This was supported by scavenger receptor-induced local increases in membrane cholesterol concentrations which generated lipid domains particularly in cell extensions and the Golgi. SR-BI was a key driver of beclin-1-dependent autophagy during acute bacterial infection of the liver and spleen. Autophagy regulated tissue infiltration of neutrophils, suppressed accumulation of Ly6C+ (inflammatory) macrophages, and prevented hepatocyte necrosis in the core of infectious foci. Perifocal levels of Ly6C+ macrophages and Ly6C− macrophages were unaffected, indicating predominant regulation of the focus core. SR-BI-triggered autophagy promoted co-elimination of apoptotic immune cells and dead bacteria but barely influenced bacterial sequestration and survival or inflammasome activation, thus exclusively counteracting damage inflicted by immune responses. Hence, SR-BI- and autophagy promote a surveillance pathway that partially responds to products of antimicrobial defenses and selectively prevents immunity-induced damage during acute infection. Our findings suggest that control of infection-associated immunopathology can be based on a unified defense operation.
Collapse
Affiliation(s)
- Susanne Pfeiler
- Institut für Laboratoriumsmedizin, Ludwig-Maximilians-Universität, Munich, 81377, Germany
| | - Avinash B Khandagale
- Institut für Laboratoriumsmedizin, Ludwig-Maximilians-Universität, Munich, 81377, Germany
| | - Astrid Magenau
- Centre for Vascular Research, ARC Centre for Excellence in Advanced Molecular Imaging and Australian Centre for Nanomedicine, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Maryana Nichols
- Institut für Laboratoriumsmedizin, Ludwig-Maximilians-Universität, Munich, 81377, Germany
| | - Harry F G Heijnen
- Laboratory of Clinical Chemistry and Haematology and Cell Microscopy Center, University Medical Center Utrecht, Utrecht, 3584CX, The Netherlands
| | - Franz Rinninger
- Universitätsklinik Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Tilman Ziegler
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, 81377, Germany
| | - Stephanie Seveau
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
| | - Sören Schubert
- Max von Pettenkofer-Institut, Ludwig-Maximilians-Universität, Munich, 80336, Germany
| | - Stefan Zahler
- Institut für Pharmazeutische Biologie, Ludwig-Maximilians-Universität, Munich, 81377, Germany
| | - Admar Verschoor
- Institut für Systemische Entzündungsforschung, Universität zu Lübeck, Lübeck, 23538, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University of Bonn, Bonn, 53127, Germany.,Department of Infectious Diseases and Immunology, UMass Medical School, Worcester, MA 01605, USA.,German Center for Neurodegenerative Diseases (DZNE), Bonn, 53127, Germany
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, 81377, Germany
| | - Katharina Gaus
- Centre for Vascular Research, ARC Centre for Excellence in Advanced Molecular Imaging and Australian Centre for Nanomedicine, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Bernd Engelmann
- Institut für Laboratoriumsmedizin, Ludwig-Maximilians-Universität, Munich, 81377, Germany
| |
Collapse
|
54
|
Yue Y, Li P, Song N, Li B, Li Z, Guo Y, Zhang W, Wei MQ, Gai Z, Meng H, Wang J, Qin L. Genomic and immunologic factors associated with viral pathogenesis in a lethal EV71 infected neonatal mouse model. Mol Med Rep 2016; 13:4183-90. [PMID: 27035332 PMCID: PMC4838153 DOI: 10.3892/mmr.2016.5080] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 03/07/2016] [Indexed: 01/01/2023] Open
Abstract
Hand, foot and mouth disease (HFMD) caused by enterovirus 71 (EV71) has emerged as a major health problem in China and worldwide. The present study aimed to understand the virological features of EV71 and host responses resulting from EV71 infection. Six different EV71 strains were isolated from HFMD patients with severe or mild clinical symptoms, and were analyzed for pathogenicity in vitro and in vivo. The results demonstrated that the six virus strains exhibited similar cytopathogenic effects on susceptible MA104 cells. However, marked differences in histological and immunopathological changes were observed when mice were inoculated with the different virus strains. Thus, the viruses studied were divided into two groups, highly or weakly pathogenic. Two representative virus strains, JN200804 and JN200803 (highly and weakly pathogenic, respectively) were studied further to investigate pathogenicity-associated factors, including genetic mutations and immunopathogenesis. The present study has demonstrated that highly pathogenic strains have stable genome and amino acid sequences. Notably, the present study demonstrated that a highly pathogenic strain induced a significant increase of the bulk CD4 T cell levels at 3 days post‑inoculation. In conclusion, the current study demonstrates that genomic and immunologic factors may be responsible for the multiple tissue damage caused by highly pathogenic EV71 infection.
Collapse
Affiliation(s)
- Yingying Yue
- Key Laboratory of Rare and Uncommon Diseases, Department of Microbiology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| | - Peng Li
- Key Laboratory of Rare and Uncommon Diseases, Department of Microbiology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| | - Nannan Song
- Key Laboratory of Rare and Uncommon Diseases, Department of Microbiology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| | - Bingqing Li
- Key Laboratory of Rare and Uncommon Diseases, Department of Microbiology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| | - Zhihui Li
- Key Laboratory of Rare and Uncommon Diseases, Department of Microbiology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| | - Yuqi Guo
- Key Laboratory of Rare and Uncommon Diseases, Department of Microbiology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| | - Weidong Zhang
- Key Laboratory of Rare and Uncommon Diseases, Department of Microbiology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| | - Ming Q Wei
- Division of Molecular and Gene Therapies, Griffith Health Institute and School of Medical Science, Griffith University, Gold Coast, Queensland 4215, Australia
| | - Zhongtao Gai
- Department of Comprehensive Internal Medicine, Jinan Children's Hospital, Jinan, Shandong 250022, P.R. China
| | - Hong Meng
- Key Laboratory of Rare and Uncommon Diseases, Department of Microbiology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| | - Jiwen Wang
- Department of Neurology, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, P.R. China
| | - Lizeng Qin
- Key Laboratory of Rare and Uncommon Diseases, Department of Microbiology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| |
Collapse
|
55
|
Bröker BM, Mrochen D, Péton V. The T Cell Response to Staphylococcus aureus. Pathogens 2016; 5:pathogens5010031. [PMID: 26999219 PMCID: PMC4810152 DOI: 10.3390/pathogens5010031] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 03/01/2016] [Accepted: 03/08/2016] [Indexed: 01/04/2023] Open
Abstract
Staphylococcus aureus (S. aureus) is a dangerous pathogen and a leading cause of both nosocomial and community acquired bacterial infection worldwide. However, on the other hand, we are all exposed to this bacterium, often within the first hours of life, and usually manage to establish equilibrium and coexist with it. What does the adaptive immune system contribute toward lifelong control of S. aureus? Will it become possible to raise or enhance protective immune memory by vaccination? While in the past the S. aureus-specific antibody response has dominated this discussion, the research community is now coming to appreciate the role that the cellular arm of adaptive immunity, the T cells, plays. There are numerous T cell subsets, each with differing functions, which together have the ability to orchestrate the immune response to S. aureus and hence to tip the balance between protection and pathology. This review summarizes the state of the art in this dynamic field of research.
Collapse
Affiliation(s)
- Barbara M Bröker
- Department of Immunology, University Medicine Greifswald, Sauerbruchstraße DZ7, 17475 Greifswald, Germany.
| | - Daniel Mrochen
- Department of Immunology, University Medicine Greifswald, Sauerbruchstraße DZ7, 17475 Greifswald, Germany.
| | - Vincent Péton
- Department of Immunology, University Medicine Greifswald, Sauerbruchstraße DZ7, 17475 Greifswald, Germany.
| |
Collapse
|
56
|
Mettu RR, Charles T, Landry SJ. CD4+ T-cell epitope prediction using antigen processing constraints. J Immunol Methods 2016; 432:72-81. [PMID: 26891811 DOI: 10.1016/j.jim.2016.02.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 12/10/2015] [Accepted: 02/11/2016] [Indexed: 02/02/2023]
Abstract
T-cell CD4+ epitopes are important targets of immunity against infectious diseases and cancer. State-of-the-art methods for MHC class II epitope prediction rely on supervised learning methods in which an implicit or explicit model of sequence specificity is constructed using a training set of peptides with experimentally tested MHC class II binding affinity. In this paper we present a novel method for CD4+ T-cell eptitope prediction based on modeling antigen-processing constraints. Previous work indicates that dominant CD4+ T-cell epitopes tend to occur adjacent to sites of initial proteolytic cleavage. Given an antigen with known three-dimensional structure, our algorithm first aggregates four types of conformational stability data in order to construct a profile of stability that allows us to identify regions of the protein that are most accessible to proteolysis. Using this profile, we then construct a profile of epitope likelihood based on the pattern of transitions from unstable to stable regions. We validate our method using 35 datasets of experimentally measured CD4+ T cell responses of mice bearing I-Ab or HLA-DR4 alleles as well as of human subjects. Overall, our results show that antigen processing constraints provide a significant source of predictive power. For epitope prediction in single-allele systems, our approach can be combined with sequence-based methods, or used in instances where little or no training data is available. In multiple-allele systems, sequence-based methods can only be used if the allele distribution of a population is known. In contrast, our approach does not make use of MHC binding prediction, and is thus agnostic to MHC class II genotypes.
Collapse
Affiliation(s)
- Ramgopal R Mettu
- Department of Computer Science, Tulane University, New Orleans, LA, USA; Vector-Borne Infectious Diseases Research Center, Tulane University, New Orleans, LA, USA.
| | - Tysheena Charles
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, USA
| | - Samuel J Landry
- Department of Biochemistry and Molecular Biology, Tulane School of Medicine, New Orleans, LA, USA
| |
Collapse
|
57
|
Sommerstein R, Flatz L, Remy MM, Malinge P, Magistrelli G, Fischer N, Sahin M, Bergthaler A, Igonet S, ter Meulen J, Rigo D, Meda P, Rabah N, Coutard B, Bowden TA, Lambert PH, Siegrist CA, Pinschewer DD. Arenavirus Glycan Shield Promotes Neutralizing Antibody Evasion and Protracted Infection. PLoS Pathog 2015; 11:e1005276. [PMID: 26587982 PMCID: PMC4654586 DOI: 10.1371/journal.ppat.1005276] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 10/22/2015] [Indexed: 01/05/2023] Open
Abstract
Arenaviruses such as Lassa virus (LASV) can cause severe hemorrhagic fever in humans. As a major impediment to vaccine development, delayed and weak neutralizing antibody (nAb) responses represent a unifying characteristic of both natural infection and all vaccine candidates tested to date. To investigate the mechanisms underlying arenavirus nAb evasion we engineered several arenavirus envelope-chimeric viruses and glycan-deficient variants thereof. We performed neutralization tests with sera from experimentally infected mice and from LASV-convalescent human patients. NAb response kinetics in mice correlated inversely with the N-linked glycan density in the arenavirus envelope protein’s globular head. Additionally and most intriguingly, infection with fully glycosylated viruses elicited antibodies, which neutralized predominantly their glycan-deficient variants, both in mice and humans. Binding studies with monoclonal antibodies indicated that envelope glycans reduced nAb on-rate, occupancy and thereby counteracted virus neutralization. In infected mice, the envelope glycan shield promoted protracted viral infection by preventing its timely elimination by the ensuing antibody response. Thus, arenavirus envelope glycosylation impairs the protective efficacy rather than the induction of nAbs, and thereby prevents efficient antibody-mediated virus control. This immune evasion mechanism imposes limitations on antibody-based vaccination and convalescent serum therapy. Neutralizing antibodies (nAbs) represent a key principle of antiviral immunity. Protective vaccines aim at inducing nAbs to prevent viral infection, and infusion of nAbs in convalescent patient serum can offer a potent antiviral therapy. Certain viruses, however, have found ways to evade nAb control. Amongst them are high-risk pathogens of the arenavirus family such as Lassa virus (LASV), which is a frequent cause of hemorrhagic fever in West Africa. Here we unveil the molecular strategy by which arenaviruses escape antibody neutralization and avoid efficient immune control. We show that their surface is decorated with sugar moieties, serving to shield the virus against the neutralizing effect of the host’s antibodies. This immune evasion strategy differs from those described for other viruses, in which sugars impair primarily the induction of antibodies or allow for viral mutational escape. The arenavirus sugar coat renders the host nAb response inefficient and as a consequence thereof, the host fails to promptly control the infection. Our results offer a compelling explanation for the long history of failures in trying to make a nAb-based vaccine against LASV or in using convalescent serum for therapy. These mechanistic insights will support vaccine development efforts against arenaviruses such as LASV.
Collapse
Affiliation(s)
- Rami Sommerstein
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- World Health Organization Collaborating Centre for Vaccine Immunology, University of Geneva, Geneva, Switzerland
| | - Lukas Flatz
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Melissa M. Remy
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | | | | | - Mehmet Sahin
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Andreas Bergthaler
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Sebastien Igonet
- Institut Pasteur, Département de Virologie, Unité de Virologie Structurale and CNRS UMR 3569 Virologie, Paris, France
| | - Jan ter Meulen
- Institute of Virology, Philipps University Marburg, Marburg, Germany
| | - Dorothée Rigo
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Nadia Rabah
- AFMB, UMR7257 CNRS/Aix Marseille Université, Marseille, France
| | - Bruno Coutard
- AFMB, UMR7257 CNRS/Aix Marseille Université, Marseille, France
| | - Thomas A. Bowden
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Paul-Henri Lambert
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- World Health Organization Collaborating Centre for Vaccine Immunology, University of Geneva, Geneva, Switzerland
| | - Claire-Anne Siegrist
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- World Health Organization Collaborating Centre for Vaccine Immunology, University of Geneva, Geneva, Switzerland
| | - Daniel D. Pinschewer
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- World Health Organization Collaborating Centre for Vaccine Immunology, University of Geneva, Geneva, Switzerland
- Division of Experimental Virology, Department of Biomedicine, University of Basel, Basel, Switzerland
- * E-mail:
| |
Collapse
|
58
|
Brown AF, Murphy AG, Lalor SJ, Leech JM, O’Keeffe KM, Mac Aogáin M, O’Halloran DP, Lacey KA, Tavakol M, Hearnden CH, Fitzgerald-Hughes D, Humphreys H, Fennell JP, van Wamel WJ, Foster TJ, Geoghegan JA, Lavelle EC, Rogers TR, McLoughlin RM. Memory Th1 Cells Are Protective in Invasive Staphylococcus aureus Infection. PLoS Pathog 2015; 11:e1005226. [PMID: 26539822 PMCID: PMC4634925 DOI: 10.1371/journal.ppat.1005226] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 09/23/2015] [Indexed: 12/15/2022] Open
Abstract
Mechanisms of protective immunity to Staphylococcus aureus infection in humans remain elusive. While the importance of cellular immunity has been shown in mice, T cell responses in humans have not been characterised. Using a murine model of recurrent S. aureus peritonitis, we demonstrated that prior exposure to S. aureus enhanced IFNγ responses upon subsequent infection, while adoptive transfer of S. aureus antigen-specific Th1 cells was protective in naïve mice. Translating these findings, we found that S. aureus antigen-specific Th1 cells were also significantly expanded during human S. aureus bloodstream infection (BSI). These Th1 cells were CD45RO+, indicative of a memory phenotype. Thus, exposure to S. aureus induces memory Th1 cells in mice and humans, identifying Th1 cells as potential S. aureus vaccine targets. Consequently, we developed a model vaccine comprising staphylococcal clumping factor A, which we demonstrate to be an effective human T cell antigen, combined with the Th1-driving adjuvant CpG. This novel Th1-inducing vaccine conferred significant protection during S. aureus infection in mice. This study notably advances our understanding of S. aureus cellular immunity, and demonstrates for the first time that a correlate of S. aureus protective immunity identified in mice may be relevant in humans. Staphylococcus aureus is a leading cause of skin, soft tissue and bone infections and, most seriously, bloodstream infection. When S. aureus does get into the bloodstream, it is more likely to kill than any other bacterial infection, despite all the treatments modern medicine has to offer. It has thus far developed resistance to all antibiotics licensed to treat it. Thus, there is an urgent need to develop a vaccine against S. aureus. However, such a vaccine remains elusive. This is largely due to the fact that we have a very limited understanding of how our immune system fights this infection. Here, we examine how certain T cells of the mouse immune system effectively recognise and respond to S. aureus, and show that transferring these cells to other mice improves their ability to clear infection. We then demonstrate that a vaccine which drives these specific T cells also improves clearance of infection. Until recently, it was not known if human T cells could recognise and respond to S. aureus. Here we show, for the first time, that these cells are expanded in patients with S. aureus bloodstream infection, and suggest that they should be targeted in anti-S. aureus vaccines.
Collapse
Affiliation(s)
- Aisling F. Brown
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Alison G. Murphy
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Stephen J. Lalor
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - John M. Leech
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Kate M. O’Keeffe
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Micheál Mac Aogáin
- Department of Clinical Microbiology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Dara P. O’Halloran
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Keenan A. Lacey
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Mehri Tavakol
- Department of Medical Microbiology & Infectious Diseases, Erasmus Medical Center, Rotterdam, Netherlands
| | - Claire H. Hearnden
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | - Hilary Humphreys
- Department of Clinical Microbiology, Royal College of Surgeons in Ireland, Dublin, Ireland
- Department of Microbiology, Beaumont Hospital, Dublin, Ireland
| | - Jérôme P. Fennell
- Department of Clinical Microbiology, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Department of Clinical Microbiology, Adelaide Meath & National Children’s Hospital, Dublin, Ireland
| | - Willem J. van Wamel
- Department of Medical Microbiology & Infectious Diseases, Erasmus Medical Center, Rotterdam, Netherlands
| | - Timothy J. Foster
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Joan A. Geoghegan
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Ed C. Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Trinity College Dublin, Dublin, Ireland
| | - Thomas R. Rogers
- Department of Clinical Microbiology, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Department of Clinical Microbiology, St. James's Hospital, Dublin, Ireland
| | - Rachel M. McLoughlin
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- * E-mail:
| |
Collapse
|
59
|
Abstract
Chronic viral infections represent a unique challenge to the infected host. Persistently replicating viruses outcompete or subvert the initial antiviral response, allowing the establishment of chronic infections that result in continuous stimulation of both the innate and adaptive immune compartments. This causes a profound reprogramming of the host immune system, including attenuation and persistent low levels of type I interferons, progressive loss (or exhaustion) of CD8(+) T cell functions, and specialization of CD4(+) T cells to produce interleukin-21 and promote antibody-mediated immunity and immune regulation. Epigenetic, transcriptional, posttranscriptional, and metabolic changes underlie this adaptation or recalibration of immune cells to the emerging new environment in order to strike an often imperfect balance between the host and the infectious pathogen. In this review we discuss the common immunological hallmarks observed across a range of different persistently replicating viruses and host species, the underlying molecular mechanisms, and the biological and clinical implications.
Collapse
Affiliation(s)
- Elina I Zuniga
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, California 92093;
| | - Monica Macal
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, California 92093;
| | - Gavin M Lewis
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, California 92093;
| | - James A Harker
- Section of Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London SW7 2AZ, United Kingdom
| |
Collapse
|
60
|
Clouthier DL, Watts TH. TNFRs and Control of Chronic LCMV Infection: Implications for Therapy. Trends Immunol 2015; 36:697-708. [PMID: 26481667 DOI: 10.1016/j.it.2015.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 09/08/2015] [Accepted: 09/15/2015] [Indexed: 12/11/2022]
Abstract
The control of persistent viral infections requires the immune system to limit the spread of the virus while avoiding immunopathology. Recent studies have revealed that members of the tumor necrosis factor receptor (TNFR) superfamily play unique and pivotal roles in control of chronic lymphocytic choriomeningitis virus (LCMV) infection and in some settings can tip the balance between immune control and immune pathology. We review these findings and discuss how our understanding of the role of TNFRs in the immune response to chronic LCMV infection may shed light on what happens during HIV infection in humans. We discuss preclinical models of TNF/TNFR family-targeted immunotherapy of chronic LCMV infection and evaluate which TNFRs present the most promising targets for immune intervention.
Collapse
Affiliation(s)
- Derek L Clouthier
- Department of Immunology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Tania H Watts
- Department of Immunology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
61
|
Jaigirdar SA, MacLeod MKL. Development and Function of Protective and Pathologic Memory CD4 T Cells. Front Immunol 2015; 6:456. [PMID: 26441961 PMCID: PMC4561815 DOI: 10.3389/fimmu.2015.00456] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 08/24/2015] [Indexed: 12/27/2022] Open
Abstract
Immunological memory is one of the defining features of the adaptive immune system. As key orchestrators and mediators of immunity, CD4 T cells are central to the vast majority of adaptive immune responses. Generated following an immune response, memory CD4 T cells retain pertinent information about their activation environment enabling them to make rapid effector responses upon reactivation. These responses can either benefit the host by hastening the control of pathogens or cause damaging immunopathology. Here, we will discuss the diversity of the memory CD4 T cell pool, the signals that influence the transition of activated T cells into that pool, and highlight how activation requirements differ between naïve and memory CD4 T cells. A greater understanding of these factors has the potential to aid the design of more effective vaccines and to improve regulation of pathologic CD4 T cells, such as in the context of autoimmunity and allergy.
Collapse
Affiliation(s)
- Shafqat Ahrar Jaigirdar
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, University of Glasgow , Glasgow , UK
| | - Megan K L MacLeod
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, University of Glasgow , Glasgow , UK
| |
Collapse
|
62
|
Penaloza-MacMaster P, Provine NM, Blass E, Barouch DH. CD4 T Cell Depletion Substantially Augments the Rescue Potential of PD-L1 Blockade for Deeply Exhausted CD8 T Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:1054-63. [PMID: 26116499 DOI: 10.4049/jimmunol.1403237] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 06/02/2015] [Indexed: 01/02/2023]
Abstract
In various models of chronic infections and cancers, blockade of the inhibitory programmed cell death-1 (PD-1) pathway has been shown to be promising at restoring immune function. However, there is not a complete understanding of the factors that influence responsiveness to programmed death-ligand 1 (PD-L1) blockade. In particular, it is currently unclear whether the efficacy of PD-L1 blockade is dependent on the stage of disease. In a model of chronic lymphocytic choriomeningitis virus infection in mice, we show that exhausted CD8 T cells during the late stage of infection are refractory to rescue by PD-L1 blockade. Interestingly, PD-L1 blockade during the late stage of infection resulted in a biased expansion of PD-1(+) CTLA-4(+) regulatory T cells (Tregs) over antiviral CD8 T cells. Although previous studies have shown that Treg ablation can enhance the immune rescue by PD-L1 blockade, this regimen may induce lethal autoimmunity. In this report, we show that PD-L1 blockade together with CD4 T cell depletion effectively rescued deeply exhausted CD8 T cells and enhanced antiviral control during the late stage of chronic infection without any associated mortality. These data demonstrate the pleiotropic effects of anti-PD-L1 therapy on both virus-specific CD8 T cells and Tregs, and suggest a novel strategy for effectively rescuing deeply exhausted CD8 T cells.
Collapse
Affiliation(s)
- Pablo Penaloza-MacMaster
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215; and
| | - Nicholas M Provine
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215; and
| | - Eryn Blass
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215; and
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215; and Ragon Institute of MGH, MIT and Harvard, Boston, MA 02114
| |
Collapse
|
63
|
Durlanik S, Thiel A. Requirement of immune system heterogeneity for protective immunity. Vaccine 2015; 33:5308-12. [PMID: 26073012 DOI: 10.1016/j.vaccine.2015.05.096] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 05/15/2015] [Accepted: 05/20/2015] [Indexed: 01/24/2023]
Abstract
Although our knowledge on the immune system and immunological memory has expanded enormously during the last decades, the development of strategies to induce robust protective memory against infections and tumors remains challenging. Intense efforts and immense resources have been put into the development of vaccines. However, effective tools to assess protective immunity, beyond neutralizing antibody titers and cytotoxic T cell activity, are still missing. Previous trials have primarily focused on individual cell subsets to induce and maintain protection while current research emphasizes the importance of functional heterogeneity and necessity of efficient communication within the immunological network. In this review, established knowledge as well as current perspectives on protective immunological memory will be discussed comprehensively.
Collapse
Affiliation(s)
- Sibel Durlanik
- Regenerative Immunology and Aging, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité University Medicine, CVK, Föhrer Str. 15, Berlin 13353, Germany.
| | - Andreas Thiel
- Regenerative Immunology and Aging, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité University Medicine, CVK, Föhrer Str. 15, Berlin 13353, Germany
| |
Collapse
|