1
|
Immune correlates of protection following Rift Valley fever virus vaccination. NPJ Vaccines 2022; 7:129. [PMID: 36307416 PMCID: PMC9616434 DOI: 10.1038/s41541-022-00551-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 10/11/2022] [Indexed: 11/27/2022] Open
Abstract
Rift Valley fever virus (RVFV) is a hemorrhagic fever virus with the potential for significant economic and public health impact. Vaccination with an attenuated strain, DelNSsRVFV, provides protection from an otherwise lethal RVFV challenge, but mechanistic determinants of protection are undefined. In this study, a murine model was used to assess the contributions of humoral and cellular immunity to DelNSsRVFV-mediated protection. Vaccinated mice depleted of T cells were protected against subsequent challenge, and passive transfer of immune serum from vaccinated animals to naïve animals was also protective, demonstrating that T cells were dispensable in the presence of humoral immunity and that humoral immunity alone was sufficient. Animals depleted of B cells and then vaccinated were protected against challenge. Total splenocytes, but not T cells alone, B cells alone, or B + T cells harvested from vaccinated animals and then transferred to naïve animals were sufficient to confer protection, suggesting that multiple cellular interactions were required for effective cellular immunity. Together, these data indicate that humoral immunity is sufficient to confer vaccine-mediated protection and suggests that cellular immunity plays a role in protection that requires the interaction of various cellular components.
Collapse
|
2
|
Chung YR, Dangi T, Palacio N, Sanchez S, Penaloza-MacMaster P. Adoptive B cell therapy for chronic viral infection. Front Immunol 2022; 13:908707. [PMID: 35958615 PMCID: PMC9361846 DOI: 10.3389/fimmu.2022.908707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
T cell-based therapies have been widely explored for the treatment of cancer and chronic infection, but B cell-based therapies have remained largely unexplored. To study the effect of B cell therapy, we adoptively transferred virus-specific B cells into mice that were chronically infected with lymphocytic choriomeningitis virus (LCMV). Adoptive transfer of virus-specific B cells resulted in increase in antibody titers and reduction of viral loads. Importantly, the efficacy of B cell therapy was partly dependent on antibody effector functions, and was improved by co-transferring virus-specific CD4 T cells. These findings provide a proof-of-concept that adoptive B cell therapy can be effective for the treatment of chronic infections, but provision of virus-specific CD4 T cells may be critical for optimal virus neutralization.
Collapse
|
3
|
Diethelm P, Schmitz I, Iten I, Kisielow J, Matsushita M, Kopf M. LCMV induced down-regulation of HVEM on anti-viral T cells is critical for an efficient effector response. Eur J Immunol 2022; 52:924-935. [PMID: 35344223 PMCID: PMC9321772 DOI: 10.1002/eji.202048569] [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: 03/04/2020] [Revised: 03/01/2022] [Accepted: 03/24/2022] [Indexed: 11/28/2022]
Abstract
T‐cell responses against tumors and pathogens are critically shaped by cosignaling molecules providing a second signal. Interaction of herpes virus entry mediator (HVEM, CD270, TNFRSF14) with multiple ligands has been proposed to promote or inhibit T‐cell responses and inflammation, dependent on the context. In this study, we show that absence of HVEM did neither affect generation of effector nor maintenance of memory antiviral T cells and accordingly viral clearance upon acute and chronic lymphocytic choriomeningitis virus (LCMV) infection, due to potent HVEM downregulation during infection. Notably, overexpression of HVEM on virus‐specific CD8+ T cells resulted in a reduction of effector cells, whereas numbers of memory cells were increased. Overall, this study indicates that downregulation of HVEM driven by LCMV infection ensures an efficient acute response at the price of impaired formation of T‐cell memory.
Collapse
Affiliation(s)
- Patrizia Diethelm
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, 8093, Switzerland
| | - Iwana Schmitz
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, 8093, Switzerland
| | - Irina Iten
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, 8093, Switzerland
| | - Jan Kisielow
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, 8093, Switzerland
| | - Mai Matsushita
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, 8093, Switzerland
| | - Manfred Kopf
- Molecular Biomedicine, Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, 8093, Switzerland
| |
Collapse
|
4
|
Reuther P, Martin K, Kreutzfeldt M, Ciancaglini M, Geier F, Calabrese D, Merkler D, Pinschewer DD. Persistent RNA virus infection is short-lived at the single-cell level but leaves transcriptomic footprints. J Exp Med 2021; 218:212556. [PMID: 34398180 PMCID: PMC8493862 DOI: 10.1084/jem.20210408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 06/14/2021] [Accepted: 07/28/2021] [Indexed: 12/14/2022] Open
Abstract
Several RNA viruses can establish life-long persistent infection in mammalian hosts, but the fate of individual virus-infected cells remains undefined. Here we used Cre recombinase-encoding lymphocytic choriomeningitis virus to establish persistent infection in fluorescent cell fate reporter mice. Virus-infected hepatocytes underwent spontaneous noncytolytic viral clearance independently of type I or type II interferon signaling or adaptive immunity. Viral clearance was accompanied by persistent transcriptomic footprints related to proliferation and extracellular matrix remodeling, immune responses, and metabolism. Substantial overlap with persistent epigenetic alterations in HCV-cured patients suggested a universal RNA virus-induced transcriptomic footprint. Cell-intrinsic clearance occurred in cell culture, too, with sequential infection, reinfection cycles separated by a period of relative refractoriness to infection. Our study reveals that systemic persistence of a prototypic noncytolytic RNA virus depends on continuous spread and reinfection. Yet undefined cell-intrinsic mechanisms prevent viral persistence at the single-cell level but give way to profound transcriptomic alterations in virus-cleared cells.
Collapse
Affiliation(s)
- Peter Reuther
- Department of Biomedicine, Division of Experimental Virology, University of Basel, Basel, Switzerland
| | - Katrin Martin
- Department of Biomedicine, Division of Experimental Virology, University of Basel, Basel, Switzerland
| | - Mario Kreutzfeldt
- Department of Pathology and Immunology, Division of Clinical Pathology, Geneva Faculty of Medicine, Geneva University and University Hospital, Geneva, Switzerland
| | - Matias Ciancaglini
- Department of Biomedicine, Division of Experimental Virology, University of Basel, Basel, Switzerland
| | - Florian Geier
- Department of Biomedicine, Bioinformatics Core Facility, University Hospital Basel, Basel, Switzerland
| | - Diego Calabrese
- Department of Biomedicine, Histology Core Facility, University Hospital Basel, Basel, Switzerland
| | - Doron Merkler
- Department of Pathology and Immunology, Division of Clinical Pathology, Geneva Faculty of Medicine, Geneva University and University Hospital, Geneva, Switzerland
| | - Daniel D Pinschewer
- Department of Biomedicine, Division of Experimental Virology, University of Basel, Basel, Switzerland
| |
Collapse
|
5
|
Vincenti I, Merkler D. New advances in immune components mediating viral control in the CNS. Curr Opin Virol 2021; 47:68-78. [PMID: 33636592 DOI: 10.1016/j.coviro.2021.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/01/2021] [Accepted: 02/05/2021] [Indexed: 11/26/2022]
Abstract
Protective immune responses in the central nervous system (CNS) must act efficiently but need to be tightly controlled to avoid excessive damage to this vital organ. Under homeostatic conditions, the immune surveillance of the CNS is mediated by innate immune cells together with subsets of memory lymphocytes accumulating over lifetime. Accordingly, a wide range of immune responses can be triggered upon pathogen infection that can be associated with devastating clinical outcomes, and which most frequently are due to neurotropic viruses. Here, we discuss recent advances about our understanding of anti-viral immune responses with special emphasis on mechanisms operating in the various anatomical compartments of the CNS.
Collapse
Affiliation(s)
- Ilena Vincenti
- University of Geneva, Department of Pathology and Immunology, Geneva, Switzerland
| | - Doron Merkler
- University of Geneva, Department of Pathology and Immunology, Geneva, Switzerland; Division of Clinical Pathology, Geneva University Hospital, 1211 Geneva, Switzerland.
| |
Collapse
|
6
|
Kräutler NJ, Yermanos A, Pedrioli A, Welten SPM, Lorgé D, Greczmiel U, Bartsch I, Scheuermann J, Kiefer JD, Eyer K, Menzel U, Greiff V, Neri D, Stadler T, Reddy ST, Oxenius A. Quantitative and Qualitative Analysis of Humoral Immunity Reveals Continued and Personalized Evolution in Chronic Viral Infection. Cell Rep 2020; 30:997-1012.e6. [PMID: 31995768 DOI: 10.1016/j.celrep.2019.12.088] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 11/20/2019] [Accepted: 12/23/2019] [Indexed: 12/31/2022] Open
Abstract
Control of established chronic lymphocytic choriomeningitis virus (LCMV) infection requires the production of neutralizing antibodies, but it remains unknown how the ensemble of antibodies evolves during ongoing infection. Here, we analyze the evolution of antibody responses during acute or chronic LCMV infection, combining quantitative functional assays and time-resolved antibody repertoire sequencing. We establish that antibody responses initially converge in both infection types on a functional and repertoire level, but diverge later during chronic infection, showing increased clonal diversity, the appearance of mouse-specific persistent clones, and distinct phylogenetic signatures. Chronic infection is characterized by a longer-lasting germinal center reaction and a continuous differentiation of plasma cells, resulting in the emergence of higher-affinity plasma cells exhibiting increased antibody secretion rates. Taken together, our findings reveal the emergence of a personalized antibody response in chronic infection and support the concept that maintaining B cell diversity throughout chronic LCMV infection correlates with the development of infection-resolving antibodies.
Collapse
Affiliation(s)
- Nike Julia Kräutler
- Institute of Microbiology, ETH Zürich, Vladimir-Prelog-Weg 1-5/10, 8093 Zürich, Switzerland
| | - Alexander Yermanos
- Institute of Microbiology, ETH Zürich, Vladimir-Prelog-Weg 1-5/10, 8093 Zürich, Switzerland; Department of Biosystems and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Alessandro Pedrioli
- Institute of Microbiology, ETH Zürich, Vladimir-Prelog-Weg 1-5/10, 8093 Zürich, Switzerland
| | - Suzanne P M Welten
- Institute of Microbiology, ETH Zürich, Vladimir-Prelog-Weg 1-5/10, 8093 Zürich, Switzerland
| | - Dominique Lorgé
- Institute of Microbiology, ETH Zürich, Vladimir-Prelog-Weg 1-5/10, 8093 Zürich, Switzerland
| | - Ute Greczmiel
- Institute of Microbiology, ETH Zürich, Vladimir-Prelog-Weg 1-5/10, 8093 Zürich, Switzerland
| | - Ilka Bartsch
- Institute of Microbiology, ETH Zürich, Vladimir-Prelog-Weg 1-5/10, 8093 Zürich, Switzerland
| | - Jörg Scheuermann
- Institute for Pharmaceutical Sciences, ETH Zürich, Vladimir-Prelog-Weg 1-5/10, 8093 Zürich, Switzerland
| | - Jonathan D Kiefer
- Institute for Pharmaceutical Sciences, ETH Zürich, Vladimir-Prelog-Weg 1-5/10, 8093 Zürich, Switzerland
| | - Klaus Eyer
- Laboratory for Functional Immune Repertoire Analysis, Institute of Pharmaceutical Sciences, D-CHAB, ETH Zürich, Zürich, Switzerland
| | - Ulrike Menzel
- Department of Biosystems and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Victor Greiff
- Department of Biosystems and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland; Department of Immunology, University of Oslo, Sognsvannsveien 20 Rikshospitalet, 0372 Oslo, Norway
| | - Dario Neri
- Institute for Pharmaceutical Sciences, ETH Zürich, Vladimir-Prelog-Weg 1-5/10, 8093 Zürich, Switzerland
| | - Tanja Stadler
- Department of Biosystems and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Sai T Reddy
- Department of Biosystems and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Annette Oxenius
- Institute of Microbiology, ETH Zürich, Vladimir-Prelog-Weg 1-5/10, 8093 Zürich, Switzerland.
| |
Collapse
|
7
|
Yermanos A, Kräutler NJ, Pedrioli A, Menzel U, Greiff V, Stadler T, Oxenius A, Reddy ST. IgM Antibody Repertoire Fingerprints in Mice Are Personalized but Robust to Viral Infection Status. Front Cell Infect Microbiol 2020; 10:254. [PMID: 32547966 PMCID: PMC7270205 DOI: 10.3389/fcimb.2020.00254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/30/2020] [Indexed: 01/11/2023] Open
Abstract
Antibody repertoire sequencing provides a molecular fingerprint of current and past pathogens encountered by the immune system. Most repertoire studies in humans require measuring the B cell response in the blood, resulting in a large bias to the IgM isotype. The extent to which the circulating IgM antibody repertoire correlates to lymphoid tissue-resident B cells in the setting of viral infection remains largely uncharacterized. Therefore, we compared the IgM repertoires from both blood and bone marrow (BM) plasma cells (PCs) following acute or chronic lymphocytic choriomeningitis virus (LCMV) infection in mice. Despite previously reported serum alterations between acute and chronic infection, IgM repertoire signatures based on clonal diversity metrics, public clones, network, and phylogenetic analysis were largely unable to distinguish infection cohorts. Our findings, however, revealed mouse-specific repertoire fingerprints between the blood and PC repertoires irrespective of infection status.
Collapse
Affiliation(s)
- Alexander Yermanos
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland.,Department of Biosystems and Engineering, ETH Zurich, Basel, Switzerland
| | | | | | - Ulrike Menzel
- Department of Biosystems and Engineering, ETH Zurich, Basel, Switzerland
| | - Victor Greiff
- Department of Immunology, University of Oslo, Oslo, Norway
| | - Tanja Stadler
- Department of Biosystems and Engineering, ETH Zurich, Basel, Switzerland
| | | | - Sai T Reddy
- Department of Biosystems and Engineering, ETH Zurich, Basel, Switzerland
| |
Collapse
|
8
|
Cooper L, Good-Jacobson KL. Dysregulation of humoral immunity in chronic infection. Immunol Cell Biol 2020; 98:456-466. [PMID: 32275789 DOI: 10.1111/imcb.12338] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/29/2020] [Accepted: 04/08/2020] [Indexed: 12/12/2022]
Abstract
Chronic viral infections disrupt the ability of the humoral immune response to produce neutralizing antibody or form effective immune memory, preventing viral clearance and making vaccine design difficult. Multiple components of the B-cell response are affected by pathogens that are not cleared from the host. Changes in the microenvironment shift production of B cells to short-lived plasma cells early in the response. Polyclonal B cells are recruited into both the plasma cell and germinal center compartments, inhibiting the formation of a targeted, high-affinity response. Finally, memory B cells shift toward an "atypical" phenotype, which may in turn result in changes to the functional properties of this population. While similar properties of B-cell dysregulation have been described across different types of persistent infections, key questions about the underlying mechanisms remain. This review will discuss the recent advances in this field, as well as highlight the critical questions about the interplay between viral load, microenvironment, the polyclonal response and atypical memory B cells that are yet to be answered. Design of new preventative treatments will rely on identifying the extrinsic and intrinsic modulators that push B cells toward an ineffective response, and thus identify new ways to guide them back onto the best path for clearance of virus and formation of effective immune memory.
Collapse
Affiliation(s)
- Lucy Cooper
- Infection and Immunity Program, The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Kim L Good-Jacobson
- Infection and Immunity Program, The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| |
Collapse
|
9
|
Zinkernagel RM. What if protective immunity is antigen-driven and not due to so-called "memory" B and T cells? Immunol Rev 2019; 283:238-246. [PMID: 29664570 DOI: 10.1111/imr.12648] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Vaccines or early childhood exposure to infection mediate immunity, that is, improved resistance against disease and death caused by a second infection with the same agent. This has been explained by and equaled to immunological memory, that is, an "altered immune system behavior" that is maintained in a presumably antigen-independent fashion. This review summarizes epidemiological and experimental data, that largely falsify this idea and that show that periodic re-exposure to antigen either, artificially as vaccines or naturally as low-level persisting antigens or infections, or immune complexes on follicular dendritic cells or endemic re-exposure is necessary for protection. Both, the huge success of vaccines in controlling childhood infections, the reduction in clinical disease and the chance of endemically re-exposure, have gradually reduced periodical re-exposure to infections and thereby endangered protective herd immunity. In parallel, vaccine deniers have created susceptibility islands even in an otherwise well vaccinated population, thereby creating a very new situation when compared to the later parts of the 20th century. If protective Immunity is-as emphasized here-antigen driven, then increasingly frequent revaccinations will be necessary (even more so with too much attenuated vaccines) to maintain both herd immunity and individual resistance to acute infections. Of course, this rule also applies to tumor vaccines.
Collapse
|
10
|
Nice TJ, Robinson BA, Van Winkle JA. The Role of Interferon in Persistent Viral Infection: Insights from Murine Norovirus. Trends Microbiol 2018; 26:510-524. [PMID: 29157967 PMCID: PMC5957778 DOI: 10.1016/j.tim.2017.10.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/20/2017] [Accepted: 10/30/2017] [Indexed: 12/21/2022]
Abstract
Persistent viral infections result from evasion or avoidance of sterilizing immunity, extend the timeframe of virus transmission, and can trigger disease. Prior studies in mouse models of persistent infection have suggested that ineffective adaptive immune responses are necessary for persistent viral infection. However, recent work in the murine norovirus (MNV) model of persistent infection demonstrates that innate immunity can control both early and persistent viral replication independently of adaptive immune effector functions. Interferons (IFNs) are central to the innate control of persistent MNV, apart from a role in modulating adaptive immunity. Furthermore, subtypes of IFN play distinct tissue-specific roles in innate control of persistent MNV infection. Type I IFN (IFN-α/β) controls systemic replication, and type III IFN (IFN-λ) controls MNV persistence in the intestinal epithelium. In this article, we review recent findings in the MNV model, highlighting the role of IFNs and innate immunity in clearing persistent viral infection, and discussing the broader implications of these findings for control of persistent human infections.
Collapse
Affiliation(s)
- Timothy J Nice
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA.
| | - Bridget A Robinson
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Jacob A Van Winkle
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
11
|
Vella LA, Herati RS, Wherry EJ. CD4 + T Cell Differentiation in Chronic Viral Infections: The Tfh Perspective. Trends Mol Med 2017; 23:1072-1087. [PMID: 29137933 PMCID: PMC5886740 DOI: 10.1016/j.molmed.2017.10.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 10/11/2017] [Accepted: 10/16/2017] [Indexed: 12/18/2022]
Abstract
CD4+ T cells play a critical role in the response to chronic viral infections during the acute phase and in the partial containment of infections once chronic infection is established. As infection persists, the virus-specific CD4+ T cell response begins to shift in phenotype. The predominant change described in both mouse and human studies of chronic viral infection is a decrease in detectable T helper type (Th)1 responses. Some Th1 loss is due to decreased proliferative potential and decreased cytokine production in the setting of chronic antigen exposure. However, recent data suggest that Th1 dysfunction is accompanied by a shift in the differentiation pathway of virus-specific CD4+ T cells, with enrichment for cells with a T follicular helper cell (Tfh) phenotype. A Tfh-like program during chronic infection has now been identified in virus-specific CD8+ T cells as well. In this review, we discuss what is known about CD4+ T cell differentiation in chronic viral infections, with a focus on the emergence of the Tfh program and the implications of this shift with respect to Tfh function and the host-pathogen interaction.
Collapse
Affiliation(s)
- Laura A Vella
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Ramin S Herati
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - E John Wherry
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
12
|
Woopen C, Straub T, Schweier O, Aichele U, Düker K, Boehm T, Pircher H. Immunological tolerance to LCMV antigens differently affects control of acute and chronic virus infection in mice. Eur J Immunol 2017; 48:120-127. [PMID: 28921501 DOI: 10.1002/eji.201747156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 07/27/2017] [Accepted: 08/09/2017] [Indexed: 11/08/2022]
Abstract
Cytotoxic T lymphocytes (CTLs) play a key role in the control of lymphocytic choriomeningitis virus (LCMV) infection. In C57BL/6 mice (H-2b ), the CTL response is mainly directed against epitopes from the LCMV glycoprotein (GP) and the nucleoprotein (NP) which represent the two major viral proteins. The role of GP- versus NP-derived epitopes for viral clearance was examined using transgenic (tg) mice ubiquitously expressing LCMV GP and NP, respectively. These mice lack GP- or NP-specific CTLs and show decreased levels of GP- or NP-specific antibodies as a result of tolerance induction. During acute LCMV infection, CTLs specific for GP- and NP-derived epitopes are generated with similar frequencies. Nonetheless, we found that lack of GP- but not of NP-specific CTLs abolished control of acute LCMV infection. In contrast, after high-dose or chronic LCMV infection, virus elimination was delayed to a similar extent in GP- and NP-tg mice. Thus, immunological tolerance to LCMV antigens differently affects virus clearance in acute and chronic infection settings. In addition, our data reveal that immunodominance of H-2b -restricted LCMV-specific CTL epitopes and their antiviral activity do not strictly correlate.
Collapse
Affiliation(s)
- Christina Woopen
- Institute for Immunology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Tobias Straub
- Institute for Immunology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Oliver Schweier
- Institute for Immunology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Ulrike Aichele
- Institute for Immunology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Katharina Düker
- Institute for Immunology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Thomas Boehm
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Hanspeter Pircher
- Institute for Immunology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
13
|
Ayala VI, Trivett MT, Barsov EV, Jain S, Piatak M, Trubey CM, Alvord WG, Chertova E, Roser JD, Smedley J, Komin A, Keele BF, Ohlen C, Ott DE. Adoptive Transfer of Engineered Rhesus Simian Immunodeficiency Virus-Specific CD8+ T Cells Reduces the Number of Transmitted/Founder Viruses Established in Rhesus Macaques. J Virol 2016; 90:9942-9952. [PMID: 27558423 PMCID: PMC5068542 DOI: 10.1128/jvi.01522-16] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 08/18/2016] [Indexed: 01/16/2023] Open
Abstract
AIDS virus infections are rarely controlled by cell-mediated immunity, in part due to viral immune evasion and immunodeficiency resulting from CD4+ T-cell infection. One likely aspect of this failure is that antiviral cellular immune responses are either absent or present at low levels during the initial establishment of infection. To test whether an extensive, timely, and effective response could reduce the establishment of infection from a high-dose inoculum, we adoptively transferred large numbers of T cells that were molecularly engineered with anti-simian immunodeficiency virus (anti-SIV) activity into rhesus macaques 3 days following an intrarectal SIV inoculation. To measure in vivo antiviral activity, we assessed the number of viruses transmitted using SIVmac239X, a molecularly tagged viral stock containing 10 genotypic variants, at a dose calculated to transmit 12 founder viruses. Single-genome sequencing of plasma virus revealed that the two animals receiving T cells expressing SIV-specific T-cell receptors (TCRs) had significantly fewer viral genotypes than the two control animals receiving non-SIV-specific T cells (means of 4.0 versus 7.5 transmitted viral genotypes; P = 0.044). Accounting for the likelihood of transmission of multiple viruses of a particular genotype, the calculated means of the total number of founder viruses transmitted were 4.5 and 14.5 in the experimental and control groups, respectively (P = 0.021). Thus, a large antiviral T-cell response timed with virus exposure can limit viral transmission. The presence of strong, preexisting T-cell responses, including those induced by vaccines, might help prevent the establishment of infection at the lower-exposure doses in humans that typically transmit only a single virus. IMPORTANCE The establishment of AIDS virus infection in an individual is essentially a race between the spreading virus and host immune defenses. Cell-mediated immune responses induced by infection or vaccination are important contributors in limiting viral replication. However, in human immunodeficiency virus (HIV)/SIV infection, the virus usually wins the race, irreversibly crippling the immune system before an effective cellular immune response is developed and active. We found that providing an accelerated response by adoptively transferring large numbers of antiviral T cells shortly after a high-dose mucosal inoculation, while not preventing infection altogether, limited the number of individual viruses transmitted. Thus, the presence of strong, preexisting T-cell responses, including those induced by vaccines, might prevent infection in humans, where the virus exposure is considerably lower.
Collapse
Affiliation(s)
- Victor I Ayala
- AIDS and Cancer Virus Program and Laboratory Animal Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Matthew T Trivett
- AIDS and Cancer Virus Program and Laboratory Animal Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Eugene V Barsov
- AIDS and Cancer Virus Program and Laboratory Animal Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Sumiti Jain
- AIDS and Cancer Virus Program and Laboratory Animal Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Michael Piatak
- AIDS and Cancer Virus Program and Laboratory Animal Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Charles M Trubey
- AIDS and Cancer Virus Program and Laboratory Animal Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - W Gregory Alvord
- DMS Applied Information & Management Sciences, Frederick National Laboratory for Cancer Research, Maryland, USA
| | - Elena Chertova
- AIDS and Cancer Virus Program and Laboratory Animal Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - James D Roser
- AIDS and Cancer Virus Program and Laboratory Animal Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Jeremy Smedley
- AIDS and Cancer Virus Program and Laboratory Animal Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Alexander Komin
- AIDS and Cancer Virus Program and Laboratory Animal Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Brandon F Keele
- AIDS and Cancer Virus Program and Laboratory Animal Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Claes Ohlen
- AIDS and Cancer Virus Program and Laboratory Animal Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - David E Ott
- AIDS and Cancer Virus Program and Laboratory Animal Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| |
Collapse
|
14
|
Hilpert C, Sitte S, Matthies A, Voehringer D. Dendritic Cells Are Dispensable for T Cell Priming and Control of Acute Lymphocytic Choriomeningitis Virus Infection. THE JOURNAL OF IMMUNOLOGY 2016; 197:2780-6. [PMID: 27549169 DOI: 10.4049/jimmunol.1502582] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 07/22/2016] [Indexed: 11/19/2022]
Abstract
Dendritic cells (DCs) are considered to be the major APCs with potent activity for priming of naive CD4 and CD8 T cells. However, T cell priming can also be achieved by other APCs including macrophages, B cells, or even nonhematopoietic cell types. Systemic low-dose infection of mice with lymphocytic choriomeningitis virus (LCMV) results in massive expansion of virus-specific CD4 and CD8 T cells. To determine the role of DCs as APCs and source of type I IFNs in this infection model, we used ΔDC mice in which DCs are constitutively ablated because of expression of the diphtheria toxin α subunit within developing DCs. ΔDC mice showed lower serum concentrations of IFN-β and IL-12p40, but normal IFN-α levels during the first days postinfection. No differences were found for proliferation of transferred TCR-transgenic cells during the early phase of infection, suggesting that T cell priming occurred with the same efficiency in wild-type and ΔDC mice. Expansion and cytokine expression of endogenous LCMV-specific T cells was comparable between wild-type and ΔDC mice during primary infection and upon rechallenge of memory mice. In both strains of infected mice the viral load was reduced below the limit of detection with the same kinetic. Further, germinal center formation and LCMV-specific Ab responses were not impaired in ΔDC mice. This indicates that DCs are dispensable as APCs for protective immunity against LCMV infection.
Collapse
Affiliation(s)
- Cornelia Hilpert
- Department of Infection Biology, University Hospital Erlangen and Friedrich Alexander University Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Selina Sitte
- Department of Infection Biology, University Hospital Erlangen and Friedrich Alexander University Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Alexander Matthies
- Department of Infection Biology, University Hospital Erlangen and Friedrich Alexander University Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - David Voehringer
- Department of Infection Biology, University Hospital Erlangen and Friedrich Alexander University Erlangen-Nuremberg, 91054 Erlangen, Germany
| |
Collapse
|
15
|
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
|
16
|
Cook KD, Shpargel KB, Starmer J, Whitfield-Larry F, Conley B, Allard DE, Rager JE, Fry RC, Davenport ML, Magnuson T, Whitmire JK, Su MA. T Follicular Helper Cell-Dependent Clearance of a Persistent Virus Infection Requires T Cell Expression of the Histone Demethylase UTX. Immunity 2015; 43:703-14. [PMID: 26431949 DOI: 10.1016/j.immuni.2015.09.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 05/14/2015] [Accepted: 08/31/2015] [Indexed: 01/01/2023]
Abstract
Epigenetic changes, including histone methylation, control T cell differentiation and memory formation, though the enzymes that mediate these processes are not clear. We show that UTX, a histone H3 lysine 27 (H3K27) demethylase, supports T follicular helper (Tfh) cell responses that are essential for B cell antibody generation and the resolution of chronic viral infections. Mice with a T cell-specific UTX deletion had fewer Tfh cells, reduced germinal center responses, lacked virus-specific immunoglobulin G (IgG), and were unable to resolve chronic lymphocytic choriomeningitis virus infections. UTX-deficient T cells showed decreased expression of interleukin-6 receptor-α and other Tfh cell-related genes that were associated with increased H3K27 methylation. Additionally, Turner Syndrome subjects, who are predisposed to chronic ear infections, had reduced UTX expression in immune cells and decreased circulating CD4(+) CXCR5(+) T cell frequency. Thus, we identify a critical link between UTX in T cells and immunity to infection.
Collapse
Affiliation(s)
- Kevin D Cook
- Department of Genetics, University of North Carolina at Chapel Hill School of Medicine, 120 Mason Farm Road, Chapel Hill, NC 27599, USA
| | - Karl B Shpargel
- Department of Genetics, University of North Carolina at Chapel Hill School of Medicine, 120 Mason Farm Road, Chapel Hill, NC 27599, USA; Carolina Center for Genome Sciences, University of North Carolina at Chapel Hill School of Medicine, 120 Mason Farm Road, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, 120 Mason Farm Road, Chapel Hill, NC 27599, USA
| | - Joshua Starmer
- Department of Genetics, University of North Carolina at Chapel Hill School of Medicine, 120 Mason Farm Road, Chapel Hill, NC 27599, USA; Carolina Center for Genome Sciences, University of North Carolina at Chapel Hill School of Medicine, 120 Mason Farm Road, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, 120 Mason Farm Road, Chapel Hill, NC 27599, USA
| | - Fatima Whitfield-Larry
- Department of Pediatrics, University of North Carolina at Chapel Hill School of Medicine, 120 Mason Farm Road, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, 120 Mason Farm Road, Chapel Hill, NC 27599, USA
| | - Bridget Conley
- Department of Pediatrics, University of North Carolina at Chapel Hill School of Medicine, 120 Mason Farm Road, Chapel Hill, NC 27599, USA
| | - Denise E Allard
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, 120 Mason Farm Road, Chapel Hill, NC 27599, USA
| | - Julia E Rager
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill School of Medicine, 120 Mason Farm Road, Chapel Hill, NC 27599, USA
| | - Rebecca C Fry
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill School of Medicine, 120 Mason Farm Road, Chapel Hill, NC 27599, USA
| | - Marsha L Davenport
- Department of Pediatrics, University of North Carolina at Chapel Hill School of Medicine, 120 Mason Farm Road, Chapel Hill, NC 27599, USA
| | - Terry Magnuson
- Department of Genetics, University of North Carolina at Chapel Hill School of Medicine, 120 Mason Farm Road, Chapel Hill, NC 27599, USA; Carolina Center for Genome Sciences, University of North Carolina at Chapel Hill School of Medicine, 120 Mason Farm Road, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, 120 Mason Farm Road, Chapel Hill, NC 27599, USA
| | - Jason K Whitmire
- Department of Genetics, University of North Carolina at Chapel Hill School of Medicine, 120 Mason Farm Road, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, 120 Mason Farm Road, Chapel Hill, NC 27599, USA.
| | - Maureen A Su
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, 120 Mason Farm Road, Chapel Hill, NC 27599, USA; Department of Pediatrics, University of North Carolina at Chapel Hill School of Medicine, 120 Mason Farm Road, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, 120 Mason Farm Road, Chapel Hill, NC 27599, USA.
| |
Collapse
|
17
|
Herz J, Johnson KR, McGavern DB. Therapeutic antiviral T cells noncytopathically clear persistently infected microglia after conversion into antigen-presenting cells. ACTA ACUST UNITED AC 2015; 212:1153-69. [PMID: 26122661 PMCID: PMC4516789 DOI: 10.1084/jem.20142047] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 06/04/2015] [Indexed: 01/12/2023]
Abstract
Clearance of neurotropic infections is challenging because the CNS is relatively intolerant of immunopathological reactions. Herz et al. use a model of persistent viral infection in mice to demonstrate therapeutic antiviral T cells can purge the CNS infection without causing tissue damage resulting from limited recruitment of inflammatory innate immune cells and conversion of microglia into APCs. Several viruses can infect the mammalian nervous system and induce neurological dysfunction. Adoptive immunotherapy is an approach that involves administration of antiviral T cells and has shown promise in clinical studies for the treatment of peripheral virus infections in humans such as cytomegalovirus (CMV), Epstein-Barr virus (EBV), and adenovirus, among others. In contrast, clearance of neurotropic infections is particularly challenging because the central nervous system (CNS) is relatively intolerant of immunopathological reactions. Therefore, it is essential to develop and mechanistically understand therapies that noncytopathically eradicate pathogens from the CNS. Here, we used mice persistently infected from birth with lymphocytic choriomeningitis virus (LCMV) to demonstrate that therapeutic antiviral T cells can completely purge the persistently infected brain without causing blood–brain barrier breakdown or tissue damage. Mechanistically, this is accomplished through a tailored release of chemoattractants that recruit antiviral T cells, but few pathogenic innate immune cells such as neutrophils and inflammatory monocytes. Upon arrival, T cells enlisted the support of nearly all brain-resident myeloid cells (microglia) by inducing proliferation and converting them into CD11c+ antigen-presenting cells (APCs). Two-photon imaging experiments revealed that antiviral CD8+ and CD4+ T cells interacted directly with CD11c+ microglia and induced STAT1 signaling but did not initiate programmed cell death. We propose that noncytopathic CNS viral clearance can be achieved by therapeutic antiviral T cells reliant on restricted chemoattractant production and interactions with apoptosis-resistant microglia.
Collapse
Affiliation(s)
- Jasmin Herz
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Kory R Johnson
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Dorian B McGavern
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
18
|
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
|
19
|
Cook KD, Kline HC, Whitmire JK. NK cells inhibit humoral immunity by reducing the abundance of CD4+ T follicular helper cells during a chronic virus infection. J Leukoc Biol 2015; 98:153-62. [PMID: 25986014 DOI: 10.1189/jlb.4hi1214-594r] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 03/31/2015] [Indexed: 01/06/2023] Open
Abstract
There is a need to understand better how to improve B cell responses and immunity to persisting virus infections, which often cause debilitating illness or death. People with chronic virus infection show evidence of improved virus control when there is a strong neutralizing antibody response, and conversely, B cell dysfunction is associated with higher viral loads. We showed previously that NK cells inhibit CD4(+) and CD8(+) T cell responses to disseminating LCMV infection and that depletion of NK cells attenuates chronic infection. Here, we examined the effect of NK cell depletion on B cell responses to LCMV infection in mice. Whereas mice infected acutely generated a peak level of antibody soon after the infection was resolved, mice infected chronically showed a continued increase in antibody levels that exceeded those after acute infection. We found that early NK cell depletion rapidly increased virus-specific antibody levels to chronic infection, and this effect depended on CD4(+) T cells and was associated with elevated numbers of CXCR5(+)CD4(+) TFH cells. However, the NK cell-depleted mice controlled the infection and by 1 mo pi, had lower TFH cell numbers and antibody levels compared with mice with sustained infection. Finally, we show that NK cell depletion improved antiviral CD8(+) T cell responses only when B cells and virus-specific antibody were present. Our data indicate that NK cells diminish immunity to chronic infection, in part, by suppressing TFH cell and antibody responses.
Collapse
Affiliation(s)
- Kevin D Cook
- *Department of Genetics and Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Hannah C Kline
- *Department of Genetics and Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Jason K Whitmire
- *Department of Genetics and Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| |
Collapse
|
20
|
Matsushita M, Freigang S, Schneider C, Conrad M, Bornkamm GW, Kopf M. T cell lipid peroxidation induces ferroptosis and prevents immunity to infection. ACTA ACUST UNITED AC 2015; 212:555-68. [PMID: 25824823 PMCID: PMC4387287 DOI: 10.1084/jem.20140857] [Citation(s) in RCA: 446] [Impact Index Per Article: 49.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 03/10/2015] [Indexed: 01/04/2023]
Abstract
Matsushita et al. investigated the role of the selenoenzyme glutathione peroxidae 4 (Gpx4) in T cell responses and found that loss of Gpx4 results in an intrinsic T cell developmental defect in the periphery, which leads to a failure to expand and protect from acute viral and parasitic infection.The defects were rescued with dietary supplementation of vitamin E. The Gp4−/− T cells accumulate membrane lipid peroxides and undergo cell death by ferroptosis. The selenoenzyme glutathione peroxidase 4 (Gpx4) is a major scavenger of phospholipid hydroperoxides. Although Gpx4 represents a key component of the reactive oxygen species-scavenging network, its relevance in the immune system is yet to be defined. Here, we investigated the importance of Gpx4 for physiological T cell responses by using T cell–specific Gpx4-deficient mice. Our results revealed that, despite normal thymic T cell development, CD8+ T cells from TΔGpx4/ΔGpx4 mice had an intrinsic defect in maintaining homeostatic balance in the periphery. Moreover, both antigen-specific CD8+ and CD4+ T cells lacking Gpx4 failed to expand and to protect from acute lymphocytic choriomeningitis virus and Leishmania major parasite infections, which were rescued with diet supplementation of high dosage of vitamin E. Notably, depletion of the Gpx4 gene in the memory phase of viral infection did not affect T cell recall responses upon secondary infection. Ex vivo, Gpx4-deficient T cells rapidly accumulated membrane lipid peroxides and concomitantly underwent cell death driven by ferroptosis but not necroptosis. These studies unveil an essential role of Gpx4 for T cell immunity.
Collapse
Affiliation(s)
- Mai Matsushita
- Molecular Biomedicine, Institute of Molecular Health Science, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Stefan Freigang
- Molecular Biomedicine, Institute of Molecular Health Science, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Christoph Schneider
- Molecular Biomedicine, Institute of Molecular Health Science, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Marcus Conrad
- Helmholtz Zentrum München, Institute of Developmental Genetics, 85764 Neuherberg, Germany
| | - Georg W Bornkamm
- Helmholtz Zentrum München, Institute of Clinical Molecular Biology and Tumor Genetics, 81377 Munich, Germany
| | - Manfred Kopf
- Molecular Biomedicine, Institute of Molecular Health Science, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
21
|
McIlwain DR, Grusdat M, Pozdeev VI, Xu HC, Shinde P, Reardon C, Hao Z, Beyer M, Bergthaler A, Häussinger D, Nolan GP, Lang KS, Lang PA. T-cell STAT3 is required for the maintenance of humoral immunity to LCMV. Eur J Immunol 2014; 45:418-27. [PMID: 25393615 PMCID: PMC4383653 DOI: 10.1002/eji.201445060] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 10/08/2014] [Accepted: 11/10/2014] [Indexed: 12/13/2022]
Abstract
STAT3 is a critical transcription factor activated downstream of cytokine signaling and is integral for the function of multiple immune cell types. Human mutations in STAT3 cause primary immunodeficiency resulting in impaired control of a variety of infections, including reactivation of latent viruses. In this study, we investigate how T-cell functions of STAT3 contribute to responses to viral infection by inducing chronic lymphocytic choriomeningitis virus (LCMV) infection in mice lacking STAT3 specifically in T cells. Although mice with conditional disruption of STAT3 in T cells were able to mount early responses to viral infection similar to control animals, including expansion of effector T cells, we found generation of T-follicular helper (Tfh) cells to be impaired. As a result, STAT3 T cell deficient mice produced attenuated germinal center reactions, and did not accumulate bone marrow virus specific IgG-secreting cells, resulting in failure to maintain levels of virus-specific IgG or mount neutralizing responses to LCMV in the serum. These effects were associated with reduced control of viral replication and prolonged infection. Our results demonstrate the importance of STAT3 in T cells for the generation of functional long-term humoral immunity to viral infections.
Collapse
Affiliation(s)
- David R McIlwain
- Department of Gastroenterology, Hepatology, and Infectious Diseases, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany; Baxter Laboratory in Stem Cell Biology, Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Perez-Shibayama C, Gil-Cruz C, Ludewig B. Plasticity and complexity of B cell responses against persisting pathogens. Immunol Lett 2014; 162:53-8. [PMID: 25068435 DOI: 10.1016/j.imlet.2014.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 07/09/2014] [Accepted: 07/17/2014] [Indexed: 11/29/2022]
Abstract
Vaccines against acute infections execute their protective effects almost exclusively via the induction of antibodies. Development of protective vaccines against persisting pathogens lags behind probably because standard immunogens and application regimen do not sufficiently stimulate those circuits in B cell activation that mediate protection. In general, B cell responses against pathogen derived-antigens are generated through complex cellular interactions requiring the coordination of innate and adaptive immune mechanisms. In this review, we summarize recent findings from prototypic infection models to exemplify how generation of protective antibodies against persisting pathogens is imprinted by particular pathogen-derived factors and how distinct CD4(+) T cell populations determine the quality of these antibodies. Clearly, it is the high plasticity of these processes that is instrumental to drive tailored B cell responses that protect the host. In sum, application of novel knowledge on B cell plasticity and complexity can guide the development of rationally designed vaccines that elicit protective antibodies against persisting pathogens.
Collapse
Affiliation(s)
- Christian Perez-Shibayama
- Institute of Immunobiology, Kantonsspital St. Gallen, Rorschacherstrasse 95, 9007 St. Gallen, Switzerland
| | - Cristina Gil-Cruz
- Institute of Immunobiology, Kantonsspital St. Gallen, Rorschacherstrasse 95, 9007 St. Gallen, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, Rorschacherstrasse 95, 9007 St. Gallen, Switzerland.
| |
Collapse
|
23
|
Xu X, Yue M, Jiang L, Deng X, Zhang Y, Zhang Y, Zhu D, Xiao W, Zhou Z, Yao W, Kong J, Yu X, Wei J. Genetic variants in human leukocyte antigen-DP influence both hepatitis C virus persistence and hepatitis C virus F protein generation in the Chinese Han population. Int J Mol Sci 2014; 15:9826-43. [PMID: 24897020 PMCID: PMC4100124 DOI: 10.3390/ijms15069826] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 05/19/2014] [Accepted: 05/21/2014] [Indexed: 12/16/2022] Open
Abstract
Chronic hepatitis C is a serious liver disease that often results in cirrhosis or hepatocellular carcinoma. The aim of this study was to assess the association of human leukocyte antigen-DP (HLA-DP) variants with risk of chronic hepatitis C virus (HCV) or anti-F antibody generation. We selected two single nucleotide polymorphisms (SNPs) in a region including HLA-DPA1 (rs3077) and HLA-DPB1 (rs9277534) and genotyped SNPs in 702 cases and 342 healthy controls from the Chinese population using TaqMan SNP genotyping assay. Moreover, the exon 2 of the HLA-DPA1 and HLA-DPB1 genes were amplified and determined by sequencing-based typing (SBT). The results showed that rs3077 significantly increased the risk of chronic HCV infection in additive models and dominant models (odds ratio (OR) = 1.32 and 1.53). The rs3077 also contributed to decrease the risk of anti-F antibody generation in additive models and dominant models (OR = 0.46 and 0.56). Subsequent analyses revealed the risk haplotypes (DPA1*0103-DPB1*0501 and DPA1*0103-DPB1*0201) and protective haplotypes (DPA1*0202-DPB1*0501 and DPA1*0202-DPB1*0202) to chronic HCV infection. Moreover, we also found that the haplotype of DPA1*0103-DPB1*0201 and DPA1*0202-DPB1*0202 were associated with the anti-F antibody generation. Our findings show that genetic variants in HLA-DP gene are associated with chronic HCV infection and anti-F antibody generation.
Collapse
Affiliation(s)
- Xiaodong Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Nanjing Medical University, Nanjing 210029, China.
| | - Ming Yue
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China.
| | - Longfeng Jiang
- Department of Infectious Diseases, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China.
| | - Xiaozhao Deng
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Nanjing Medical University, Nanjing 210029, China.
| | - Yongxiang Zhang
- Department of Infectious Diseases, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China.
| | - Yun Zhang
- Institute of Disease Control and Prevention, Huadong Research Institute for Medicine and Biotechnics, Nanjing 210002, China.
| | - Danyan Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Nanjing Medical University, Nanjing 210029, China.
| | - Wen Xiao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China.
| | - Zhenxian Zhou
- Department of Clinical Laboratory, Nanjing Second Hospital, Nanjing 210003, China.
| | - Wenjuan Yao
- Department of Pharmacology, Nantong University Medical College, Nantong 226019, China.
| | - Jing Kong
- School of Life Science and Chemical Engineering, Huaiyin Institute of Technology, Huaian 223003, China.
| | - Xiaojie Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Nanjing Medical University, Nanjing 210029, China.
| | - Juan Wei
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
24
|
Park HL, Kim YJ, Na HN, Park MY, Kim JY, Yun CW, Nam JH. IK induced by coxsackievirus B3 infection transiently downregulates expression of MHC class II through increasing cAMP. Viral Immunol 2013; 26:13-24. [PMID: 23409929 DOI: 10.1089/vim.2012.0054] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Major histocompatibility complex (MHC) class II expression is critical for the presentation of antigens in the immune response to viral infection. Consequently, some viruses regulate the MHC class II-mediated presentation of viral antigens as a mechanism of immune escape. In this study, we found that Coxsackievirus B3 (CVB3) infection transiently increased IK expression, which reduced the expression of MHC class II (I-A/I-E) on splenic B cells. Interestingly, CVB3-induced IK elevated cAMP, a downstream molecule of the G protein-coupled receptors, which inhibited MHC class II presentation on B cells. Transgenic mice expressing truncated IK showed lower expression of MHC class II on B cells than did wild-type mice after CVB3 infection. Taken together, these results imply that IK plays a role in downregulating MHC class II expression on B cells during CVB3 infection through the induction of cAMP.
Collapse
Affiliation(s)
- Hye-Lim Park
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Gyeonggi-do, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
25
|
Straub T, Schweier O, Bruns M, Nimmerjahn F, Waisman A, Pircher H. Nucleoprotein-specific nonneutralizing antibodies speed up LCMV elimination independently of complement and FcγR. Eur J Immunol 2013; 43:2338-48. [PMID: 23749409 DOI: 10.1002/eji.201343565] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 05/21/2013] [Accepted: 06/04/2013] [Indexed: 12/12/2022]
Abstract
CD8(+) T cells have an essential role in controlling lymphocytic choriomeningitis virus (LCMV) infection in mice. Here, we examined the contribution of humoral immunity, including nonneutralizing antibodies (Abs), in this infection induced by low virus inoculation doses. Mice with impaired humoral immunity readily terminated infection with the slowly replicating LCMV strain Armstrong but showed delayed virus elimination after inoculation with the faster replicating LCMV strain WE and failed to clear the rapidly replicating LCMV strain Docile, which is in contrast to the results obtained with wild-type mice. Thus, the requirement for adaptive humoral immunity to control the infection was dependent on the replication speed of the LCMV strains used. Ab transfers further showed that LCMV-specific IgG Abs isolated from LCMV immune serum accelerated virus elimination. These Abs were mainly directed against the viral nucleoprotein (NP) and completely lacked virus neutralizing activity. Moreover, mAbs specific for the LCMV NP were also able to decrease viral titers after transfer into infected hosts. Intriguingly, neither C3 nor Fcγ receptors were required for the antiviral activity of the transferred Abs. In conclusion, our study suggests that rapidly generated nonneutralizing Abs specific for the viral NP speed up virus elimination and thereby may counteract T-cell exhaustion.
Collapse
Affiliation(s)
- Tobias Straub
- Department of Immunology, Institute of Medical Microbiology and Hygiene, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | | | | | | | | | | |
Collapse
|
26
|
Khanolkar A, Williams MA, Harty JT. Antigen experience shapes phenotype and function of memory Th1 cells. PLoS One 2013; 8:e65234. [PMID: 23762323 PMCID: PMC3676405 DOI: 10.1371/journal.pone.0065234] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 04/23/2013] [Indexed: 12/22/2022] Open
Abstract
Primary and secondary (boosted) memory CD8 T cells exhibit differences in gene expression, phenotype and function. The impact of repeated antigen stimulations on memory CD4 T cells is largely unknown. To address this issue, we utilized LCMV and Listeria monocytogenes infection of mice to characterize primary and secondary antigen (Ag)-specific Th1 CD4 T cell responses. Ag-specific primary memory CD4 T cells display a CD62LloCCR7hi CD27hi CD127hi phenotype and are polyfunctional (most produce IFNγ, TNFα and IL-2). Following homologous prime-boost immunization we observed pathogen-specific differences in the rate of CD62L and CCR7 upregulation on memory CD4 T cells as well as in IL-2+IFNγco-production by secondary effectors. Phenotypic and functional plasticity of memory Th1 cells was observed following heterologous prime-boost immunization, wherein secondary memory CD4 T cells acquired phenotypic and functional characteristics dictated by the boosting agent rather than the primary immunizing agent. Our data also demonstrate that secondary memory Th1 cells accelerated neutralizing Ab formation in response to LCMV infection, suggesting enhanced capacity of this population to provide quality help for antibody production. Collectively these data have important implications for prime-boost vaccination strategies that seek to enhance protective immune responses mediated by Th1 CD4 T cell responses.
Collapse
Affiliation(s)
- Aaruni Khanolkar
- Department of Microbiology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Matthew A. Williams
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
- * E-mail: (JTH); (MAW)
| | - John T. Harty
- Department of Microbiology, University of Iowa, Iowa City, Iowa, United States of America
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
- * E-mail: (JTH); (MAW)
| |
Collapse
|
27
|
Walton S, Mandaric S, Oxenius A. CD4 T cell responses in latent and chronic viral infections. Front Immunol 2013; 4:105. [PMID: 23717308 PMCID: PMC3651995 DOI: 10.3389/fimmu.2013.00105] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 04/22/2013] [Indexed: 12/24/2022] Open
Abstract
The spectrum of tasks which is fulfilled by CD4 T cells in the setting of viral infections is large, ranging from support of CD8 T cells and humoral immunity to exertion of direct antiviral effector functions. While our knowledge about the differentiation pathways, plasticity, and memory of CD4 T cell responses upon acute infections or immunizations has significantly increased during the past years, much less is still known about CD4 T cell differentiation and their beneficial or pathological functions during persistent viral infections. In this review we summarize current knowledge about the differentiation, direct or indirect antiviral effector functions, and the regulation of virus-specific CD4 T cells in the setting of persistent latent or active chronic viral infections with a particular emphasis on herpes virus infections for the former and chronic lymphocytic choriomeningitis virus infection for the latter.
Collapse
Affiliation(s)
- Senta Walton
- Department of Microbiology and Immunology, School of Pathology and Laboratory Medicine, University of Western Australia Nedlands, WA, Australia
| | | | | |
Collapse
|
28
|
Alternative serotype adenovirus vaccine vectors elicit memory T cells with enhanced anamnestic capacity compared to Ad5 vectors. J Virol 2012; 87:1373-84. [PMID: 23152535 DOI: 10.1128/jvi.02058-12] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The failure of the adenovirus serotype 5 (Ad5) vector-based human immunodeficiency virus type 1 (HIV-1) vaccine in the STEP study has led to the development of adenovirus vectors derived from alternative serotypes, such as Ad26, Ad35, and Ad48. We have recently demonstrated that vaccines using alternative-serotype Ad vectors confer partial protection against stringent simian immunodeficiency virus (SIV) challenges in rhesus monkeys. However, phenotypic differences between the T cell responses elicited by Ad5 and those of alternative-serotype Ad vectors remain unexplored. Here, we report the magnitude, phenotype, functionality, and recall capacity of memory T cell responses elicited in mice by Ad5, Ad26, Ad35, and Ad48 vectors expressing lymphocytic choriomeningitis virus (LCMV) glycoprotein (GP). Our data demonstrate that memory T cells elicited by Ad5 vectors were high in magnitude but exhibited functional exhaustion and decreased anamnestic potential following secondary antigen challenge compared to Ad26, Ad35, and Ad48 vectors. These data suggest that vaccination with alternative-serotype Ad vectors offers substantial immunological advantages over Ad5 vectors, in addition to circumventing high baseline Ad5-specific neutralizing antibody titers.
Collapse
|
29
|
Yousef S, Planas R, Chakroun K, Hoffmeister-Ullerich S, Binder TMC, Eiermann TH, Martin R, Sospedra M. TCR Bias and HLA Cross-Restriction Are Strategies of Human Brain-Infiltrating JC Virus-Specific CD4+T Cells during Viral Infection. THE JOURNAL OF IMMUNOLOGY 2012; 189:3618-30. [DOI: 10.4049/jimmunol.1201612] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
30
|
Franceschini D, Del Porto P, Piconese S, Trella E, Accapezzato D, Paroli M, Morrone S, Piccolella E, Spada E, Mele A, Sidney J, Sette A, Barnaba V. Polyfunctional type-1, -2, and -17 CD8⁺ T cell responses to apoptotic self-antigens correlate with the chronic evolution of hepatitis C virus infection. PLoS Pathog 2012; 8:e1002759. [PMID: 22737070 PMCID: PMC3380931 DOI: 10.1371/journal.ppat.1002759] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Accepted: 05/03/2012] [Indexed: 12/17/2022] Open
Abstract
Caspase-dependent cleavage of antigens associated with apoptotic cells plays a prominent role in the generation of CD8⁺ T cell responses in various infectious diseases. We found that the emergence of a large population of autoreactive CD8⁺ T effector cells specific for apoptotic T cell-associated self-epitopes exceeds the antiviral responses in patients with acute hepatitis C virus infection. Importantly, they endow mixed polyfunctional type-1, type-2 and type-17 responses and correlate with the chronic progression of infection. This evolution is related to the selection of autoreactive CD8⁺ T cells with higher T cell receptor avidity, whereas those with lower avidity undergo prompt contraction in patients who clear infection. These findings demonstrate a previously undescribed strict link between the emergence of high frequencies of mixed autoreactive CD8⁺ T cells producing a broad array of cytokines (IFN-γ, IL-17, IL-4, IL-2…) and the progression toward chronic disease in a human model of acute infection.
Collapse
Affiliation(s)
- Debora Franceschini
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Paola Del Porto
- Dipartimento di Biologia e Biotecnologie “Charles Darwin”, Sapienza Università di Roma, Rome, Italy
| | - Silvia Piconese
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Emanuele Trella
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Daniele Accapezzato
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Marino Paroli
- Dipartimento di Scienze e Biotecnologie Medico-Chirurgiche, Sapienza Università di Roma, Rome, Italy
| | - Stefania Morrone
- Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, Rome, Italy
| | - Enza Piccolella
- Dipartimento di Biologia e Biotecnologie “Charles Darwin”, Sapienza Università di Roma, Rome, Italy
| | - Enea Spada
- National Centre of Epidemiology, Surveillance and Health Promotion, Istituto Superiore di Sanità, Rome, Italy
| | - Alfonso Mele
- National Centre of Epidemiology, Surveillance and Health Promotion, Istituto Superiore di Sanità, Rome, Italy
| | - John Sidney
- La Jolla Institute for Allergy and Immunology, San Diego, California, United States of America
| | - Alessandro Sette
- La Jolla Institute for Allergy and Immunology, San Diego, California, United States of America
| | - Vincenzo Barnaba
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
- Istituto Pasteur - Fondazione Cenci Bolognetti, Rome, Italy
- Fondazione Andrea Cesalpino, Rome, Italy
- * E-mail:
| |
Collapse
|
31
|
Walsh KB, Teijaro JR, Zuniga EI, Welch MJ, Fremgen DM, Blackburn SD, von Tiehl K, Wherry EJ, Flavell RA, Oldstone MBA. Toll-like receptor 7 is required for effective adaptive immune responses that prevent persistent virus infection. Cell Host Microbe 2012; 11:643-53. [PMID: 22704624 PMCID: PMC3377981 DOI: 10.1016/j.chom.2012.04.016] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2011] [Revised: 02/15/2012] [Accepted: 04/05/2012] [Indexed: 10/28/2022]
Abstract
TLR7 is an innate signaling receptor that recognizes single-stranded viral RNA and is activated by viruses that cause persistent infections. We show that TLR7 signaling dictates either clearance or establishment of life-long chronic infection by lymphocytic choriomeningitis virus (LCMV) Cl 13 but does not affect clearance of the acute LCMV Armstrong 53b strain. TLR7(-/-) mice infected with LCMV Cl 13 remained viremic throughout life from defects in the adaptive antiviral immune response-notably, diminished T cell function, exacerbated T cell exhaustion, decreased plasma cell maturation, and negligible antiviral antibody production. Adoptive transfer of TLR7(+/+) LCMV immune memory cells that enhanced clearance of persistent LCMV Cl 13 infection in TLR7(+/+) mice failed to purge LCMV Cl 13 infection in TLR7(-/-) mice, demonstrating that a TLR7-deficient environment renders antiviral responses ineffective. Therefore, methods that promote TLR7 signaling are promising treatment strategies for chronic viral infections.
Collapse
Affiliation(s)
- Kevin B. Walsh
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - John R. Teijaro
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Elina I. Zuniga
- Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Megan J. Welch
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Daniel M. Fremgen
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Shawn D. Blackburn
- Department of Microbiology and Institute for Immunology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Karl von Tiehl
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - E. John Wherry
- Department of Microbiology and Institute for Immunology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | - Michael B. A. Oldstone
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
32
|
Zinkernagel RM. Immunological memory ≠ protective immunity. Cell Mol Life Sci 2012; 69:1635-40. [PMID: 22481438 PMCID: PMC11114992 DOI: 10.1007/s00018-012-0972-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2012] [Revised: 03/13/2012] [Accepted: 03/13/2012] [Indexed: 01/12/2023]
Abstract
So-called 'immunological memory' is, in my view, a typical example where a field of enquiry, i.e. to understand long-term protection to survive reexposure to infection, has been overtaken by 'l'art pour l'art' of 'basic immunology'. The aim of this critical review is to point out some key differences between academic text book-defined immunological memory and protective immunity as viewed from a co-evolutionary point of view, both from the host and the infectious agents. A key conclusion is that 'immunological memory' of course exists, but only in particular experimental laboratory models measuring 'quicker and better' responses after an earlier immunization. These often do correlate with, but are not the key mechanisms of, protection. Protection depends on pre-existing neutralizing antibodies or pre-activated T cells at the time of infection-as documented by the importance of maternal antibodies around birth for survival of the offspring. Importantly, both high levels of antibodies and of activated T cells are antigen driven. This conclusion has serious implications for our thinking about vaccines and maintaining a level of protection in the population to deal with old and new infectious diseases.
Collapse
|
33
|
Cole DK, Gallagher K, Lemercier B, Holland CJ, Junaid S, Hindley JP, Wynn KK, Gostick E, Sewell AK, Gallimore AM, Ladell K, Price DA, Gougeon ML, Godkin A. Modification of the carboxy-terminal flanking region of a universal influenza epitope alters CD4⁺ T-cell repertoire selection. Nat Commun 2012; 3:665. [PMID: 22314361 PMCID: PMC3293629 DOI: 10.1038/ncomms1665] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Accepted: 01/05/2012] [Indexed: 02/01/2023] Open
Abstract
Human CD4(+) αβ T cells are activated via T-cell receptor recognition of peptide epitopes presented by major histocompatibility complex (MHC) class II (MHC-II). The open ends of the MHC-II binding groove allow peptide epitopes to extend beyond a central nonamer core region at both the amino- and carboxy-terminus. We have previously found that these non-bound C-terminal residues can alter T cell activation in an MHC allele-transcending fashion, although the mechanism for this effect remained unclear. Here we show that modification of the C-terminal peptide-flanking region of an influenza hemagglutinin (HA(305-320)) epitope can alter T-cell receptor binding affinity, T-cell activation and repertoire selection of influenza-specific CD4(+) T cells expanded from peripheral blood. These data provide the first demonstration that changes in the C-terminus of the peptide-flanking region can substantially alter T-cell receptor binding affinity, and indicate a mechanism through which peptide flanking residues could influence repertoire selection.
Collapse
Affiliation(s)
- David K. Cole
- Institute of Infection and Immunity, Cardiff University School of Medicine, The Henry Wellcome Building, Cardiff CF14 4XN, Wales, UK
- These authors contributed equally to this work
| | - Kathleen Gallagher
- Institute of Infection and Immunity, Cardiff University School of Medicine, The Henry Wellcome Building, Cardiff CF14 4XN, Wales, UK
- These authors contributed equally to this work
| | - Brigitte Lemercier
- Institut Pasteur, Antiviral Immunity, Biotherapy and Vaccine Unit, Department of Infection and Epidemiology, rue du Dr. Roux, 75015 Paris, France
- These authors contributed equally to this work
| | - Christopher J. Holland
- Institute of Infection and Immunity, Cardiff University School of Medicine, The Henry Wellcome Building, Cardiff CF14 4XN, Wales, UK
| | - Sayed Junaid
- Institute of Infection and Immunity, Cardiff University School of Medicine, The Henry Wellcome Building, Cardiff CF14 4XN, Wales, UK
| | - James P. Hindley
- Institute of Infection and Immunity, Cardiff University School of Medicine, The Henry Wellcome Building, Cardiff CF14 4XN, Wales, UK
| | - Katherine K. Wynn
- Institute of Infection and Immunity, Cardiff University School of Medicine, The Henry Wellcome Building, Cardiff CF14 4XN, Wales, UK
| | - Emma Gostick
- Institute of Infection and Immunity, Cardiff University School of Medicine, The Henry Wellcome Building, Cardiff CF14 4XN, Wales, UK
| | - Andrew K. Sewell
- Institute of Infection and Immunity, Cardiff University School of Medicine, The Henry Wellcome Building, Cardiff CF14 4XN, Wales, UK
| | - Awen M. Gallimore
- Institute of Infection and Immunity, Cardiff University School of Medicine, The Henry Wellcome Building, Cardiff CF14 4XN, Wales, UK
| | - Kristin Ladell
- Institute of Infection and Immunity, Cardiff University School of Medicine, The Henry Wellcome Building, Cardiff CF14 4XN, Wales, UK
| | - David A. Price
- Institute of Infection and Immunity, Cardiff University School of Medicine, The Henry Wellcome Building, Cardiff CF14 4XN, Wales, UK
| | - Marie-Lise Gougeon
- Institut Pasteur, Antiviral Immunity, Biotherapy and Vaccine Unit, Department of Infection and Epidemiology, rue du Dr. Roux, 75015 Paris, France
| | - Andrew Godkin
- Institute of Infection and Immunity, Cardiff University School of Medicine, The Henry Wellcome Building, Cardiff CF14 4XN, Wales, UK
- Department of Medicine, University Hospital of Wales, Cardiff CF14 4XW, Wales, UK
| |
Collapse
|
34
|
Audigé A, Hofer U, Dittmer U, van den Broek M, Speck RF. Evaluation of the immunomodulatory and antiviral effects of the cytokine combination IFN-α and IL-7 in the lymphocytic choriomeningitis virus and Friend retrovirus mouse infection models. Viral Immunol 2011; 24:375-85. [PMID: 21929334 DOI: 10.1089/vim.2011.0006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Existing therapies for chronic viral infections are still suboptimal or have considerable side effects, so new therapeutic strategies need to be developed. One option is to boost the host's immune response with cytokines. We have recently shown in an acute ex vivo HIV infection model that co-administration of interferon (IFN)-α and interleukin (IL)-7 allows us to combine the potent anti-HIV activity of IFN-α with the beneficial effects of IL-7 on T-cell survival and function. Here we evaluated the effect of combining IFN-α and IL-7 on viral replication in vivo in the chronic lymphocytic choriomeningitis virus (LCMV) and acute Friend retrovirus (FV) infection models. In the chronic LCMV model, cytokine treatment was started during the early replication phase (i.e., on day 7 post-infection [pi]). Under the experimental conditions used, exogenous IFN-α inhibited FV replication, but had no effect on viral replication in the LCMV model. There was no therapeutic benefit of IL-7 either alone or in combination with IFN-α in either of the two infection models. In the LCMV model, dose-dependent effects of the cytokine combination on T-cell phenotype/function were observed. It is possible that these effects would translate into antiviral activity in re-challenged mice. It is also possible that another type of IFN-α/β or induction of endogenous IFN-α/β alone or in combination with IL-7 would have antiviral activity in the LCMV model. Furthermore, we cannot exclude that some effect on viral titers would have been seen at later time points not investigated here (i.e., beyond day 34 pi). Finally, IFN-α/IL-7 may inhibit the replication of other viruses. Thus it might be worth testing these cytokines in other in vivo models of chronic viral infections.
Collapse
Affiliation(s)
- Annette Audigé
- Division of Infectious Diseases and Hospital Epidemiology, University of Zurich, Zurich, Switzerland.
| | | | | | | | | |
Collapse
|
35
|
Holst PJ, Christensen JP, Thomsen AR. Vaccination against lymphocytic choriomeningitis virus infection in MHC class II-deficient mice. THE JOURNAL OF IMMUNOLOGY 2011; 186:3997-4007. [PMID: 21357263 DOI: 10.4049/jimmunol.1001251] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The impact of prophylactic vaccination against acute and chronic infection in a Th-deficient host has not been adequately addressed because of difficulties in generating protective immunity in the absence of CD4(+) T cell help. In this study, we demonstrated that a broad CD8(+) T cell immune response could be elicited in MHC class II-deficient mice by vaccination with adenovirus encoding lymphocytic choriomeningitis virus (LCMV) glycoprotein tethered to MHC class II-associated invariant chain. Moreover, the response induced conferred significant cytolytic CD8(+) T cell-mediated protection against challenge with a high dose of the invasive clone 13 strain of LCMV. In contrast, vaccination with adenovirus encoding unlinked LCMV glycoprotein induced weak virus control in the absence of CD4(+) T cells, and mice may die of increased immunopathology associated with incomplete protection. Acute mortality was not observed in any vaccinated mice following infection with the less-invasive Traub strain. However, LCMV Traub infection caused accelerated late mortality in unvaccinated MHC class II-deficient mice; in this case, we observed a strong trend toward delayed mortality in vaccinated mice, irrespective of the nature of the vaccine. These results indicated that optimized vaccination may lead to efficient protection against acute viral infection, even in Th-deficient individuals, but that the duration of such immunity is limited. Nevertheless, for select immunodeficiencies in which CD4(+) T cell deficiency is incomplete or transient, these results are very encouraging.
Collapse
Affiliation(s)
- Peter Johannes Holst
- Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen DK-2200, Denmark
| | | | | |
Collapse
|
36
|
Characterization of the specific CD4+ T cell response against the F protein during chronic hepatitis C virus infection. PLoS One 2010; 5:e14237. [PMID: 21151917 PMCID: PMC2997803 DOI: 10.1371/journal.pone.0014237] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Accepted: 11/06/2010] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The hepatitis C virus (HCV) Alternate Reading Frame Protein (ARFP or F protein) presents a double-frame shift product of the HCV core gene. We and others have previously reported that the specific antibodies against the F protein could be raised in the sera of HCV chronically infected patients. However, the specific CD4(+) T cell responses against the F protein during HCV infection and the pathological implications remained unclear. In the current study, we screened the MHC class II-presenting epitopes of the F protein through HLA-transgenic mouse models and eventually validated the specific CD4(+) T cell responses in HCV chronically infected patients. METHODOLOGY DNA vaccination in HLA-DR1 and-DP4 transgenic mouse models, proliferation assay to test the F protein specific T cell response, genotyping of Chronic HCV patients and testing the F-peptide stimulated T cell response in the peripheral blood mononuclear cell (PBMC) by in vitro expansion and interferon (IFN)- γ intracellular staining. PRINCIPAL FINDINGS At least three peptides within HCV F protein were identified as HLA-DR or HLA-DP4 presenting epitopes by the proliferation assays in mouse models. Further study with human PBMCs evidenced the specific CD4(+) T cell responses against HCV F protein as well in patients chronically infected with HCV. CONCLUSION The current study provided the evidence for the first time that HCV F protein could elicit specific CD4(+) T cell response, which may provide an insight into the immunopathogenesis during HCV chronic infection.
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW The only unequivocal correlate of protection against primate immunodeficiency virus infection is the presence of neutralizing antibody at sufficient titre. This correlate has been determined experimentally using animal models, and the data are reproducible and robust. Recent advances have added further depth to this knowledge by moving us closer to understanding how antibodies neutralize HIV-1, and what effects they may have in vivo with regard to protection from infection and disease. RECENT FINDINGS This review will cover recent advances in our understanding of the structural basis of HIV-1 neutralization by antibody and how this understanding may relate to vaccine design, and incorporate this into the broader context of how antibodies may influence viral transmission, replication and disease. SUMMARY The sum of these findings provides a strong rationale for designing an HIV-1 vaccine on the principle of induction of neutralizing antibodies, although other effector functions of antibodies such as complement and antibody-mediated cellular immunity should also be borne in mind, as should CD4 and CD8 T cell responses.
Collapse
|
38
|
Impaired antibody response causes persistence of prototypic T cell-contained virus. PLoS Biol 2009; 7:e1000080. [PMID: 19355789 PMCID: PMC2672599 DOI: 10.1371/journal.pbio.1000080] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2008] [Accepted: 02/24/2009] [Indexed: 11/19/2022] Open
Abstract
CD8 T cells are recognized key players in control of persistent virus infections, but increasing evidence suggests that assistance from other immune mediators is also needed. Here, we investigated whether specific antibody responses contribute to control of lymphocytic choriomeningitis virus (LCMV), a prototypic mouse model of systemic persistent infection. Mice expressing transgenic B cell receptors of LCMV-unrelated specificity, and mice unable to produce soluble immunoglobulin M (IgM) exhibited protracted viremia or failed to resolve LCMV. Virus control depended on immunoglobulin class switch, but neither on complement cascades nor on Fc receptor gamma chain or Fc gamma receptor IIB. Cessation of viremia concurred with the emergence of viral envelope-specific antibodies, rather than with neutralizing serum activity, and even early nonneutralizing IgM impeded viral persistence. This important role for virus-specific antibodies may be similarly underappreciated in other primarily T cell-controlled infections such as HIV and hepatitis C virus, and we suggest this contribution of antibodies be given consideration in future strategies for vaccination and immunotherapy.
Collapse
|
39
|
Yoshida A, Tanaka R, Kodama A, Yamamoto N, Ansari AA, Tanaka Y. Identification of HIV-1 epitopes that induce the synthesis of a R5 HIV-1 suppression factor by human CD4+ T cells isolated from HIV-1 immunized hu-PBL SCID mice. Clin Dev Immunol 2009; 12:235-42. [PMID: 16584108 PMCID: PMC2270741 DOI: 10.1080/17402520500391557] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
We have previously reported that immunization of the severe combined immunodeficiency (SCID) mice reconstituted with human peripheral blood mononuclear cells (PBMC) (hu-PBL-SCID mice) with inactivated human immunodeficiency virus type-1 (HIV-1)-pulsed-autologous dendritic cells (HIV-DC) elicits HIV-1-reactive CD4(+) T cells that produce an as yet to be defined novel soluble factor in vitro with anti-viral properties against CCR5 tropic (R5) HIV-1 infection. These findings led us to perform studies designed to identify the lineage of the cell that synthesizes such a factor in vivo and define the epitopes of HIV-1 protein that have specificity for the induction of such anti-viral factor. Results of our studies show that this property is a function of CD4(+) but not CD8(+) T cells. Human CD4(+) T cells were thus recovered from the HIV-DC-immunized hu-PBL-SCID mice and were re-stimulated in vitro by co-culture for 2 days with autologous adherent PBMC as antigen presenting cells, APC previously pulsed with inactivated HIV in IL-2-containing medium to expand HIV-1-reactive CD4(+) T cells. Aliquots of these re-stimulated CD4(+) T cells were then co-cultured with similar APC's that were previously pulsed with 10 microg/ml of a panel of HIV peptides for an additional 2 days, and their culture supernatants were examined for the production of both the R5 HIV-1 suppression factor and IFN-gamma. The data presented herein show that the HIV-1 primed CD4(+) T cells produced the R5 suppression factor in response to a wide variety of HIV-1 gag, env, pol, nef or vif peptides, depending on the donor of the CD4(+) T cells. Simultaneous production of human interferon (IFN)-gamma was observed in some cases. These results indicate that human CD4(+) T cells in PBMC of HIV-1 naive donors have a wide variety of HIV-1 epitope-specific CD4(+) T cell precursors that are capable of producing the R5 HIV-1 suppression factor upon DC-based vaccination with whole inactivated HIV-1.
Collapse
Affiliation(s)
- Atsushi Yoshida
- Department of Immunology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Okinawa 903-0215, Japan
| | | | | | | | | | | |
Collapse
|
40
|
Efficient Modulation of T-cell Response by Dual-mode, Single-carrier Delivery of Cytokine-targeted siRNA and DNA Vaccine to Antigen-presenting Cells. Mol Ther 2008; 16:2011-21. [DOI: 10.1038/mt.2008.206] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
41
|
Matter MS, Claus C, Ochsenbein AF. CD4+ T cell help improves CD8+ T cell memory by retained CD27 expression. Eur J Immunol 2008; 38:1847-56. [PMID: 18506879 DOI: 10.1002/eji.200737824] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
CD4+ T cell help during the priming of CD8+ T lymphocytes imprints the capacity for optimal secondary expansion upon re-encounter with antigen. Helped memory CD8+ T cells rapidly expand in response to a secondary antigen exposure, even in the absence of T cell help and, are most efficient in protection against a re-infection. In contrast, helpless memory CTL can mediate effector function, but secondary expansion is reduced. How CD4+ T cells instruct CD8+ memory T cells during priming to undergo efficient secondary expansion has not been resolved in detail. Here, we show that memory CTL after infection with lymphocytic choriomeningitis virus are CD27(high) whereas memory CTL primed in the absence of CD4+ T cell have a reduced expression of CD27. Helpless memory CTL produced low amounts of IL-2 and did not efficiently expand after restimulation with peptide in vitro. Blocking experiments with monoclonal antibodies and the use of CD27(-/-) memory CTL revealed that CD27 ligation during restimulation increased autocrine IL-2 production and secondary expansion. Therefore, regulating CD27 expression on memory CTL is a novel mechanism how CD4+ T cells control CTL memory.
Collapse
Affiliation(s)
- Matthias S Matter
- Tumor Immunology, Department of Clinical Research, University of Berne, Bern, Switzerland
| | | | | |
Collapse
|
42
|
Truong P, McGavern DB. A novel virus carrier state to evaluate immunotherapeutic regimens: regulatory T cells modulate the pathogenicity of antiviral memory cells. THE JOURNAL OF IMMUNOLOGY 2008; 181:1161-9. [PMID: 18606669 DOI: 10.4049/jimmunol.181.2.1161] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Restrictions in the diversity of an adaptive immune repertoire can facilitate viral persistence. Because a host afflicted with an immune deficiency is not likely to purge a persistent infection using endogenous mechanisms, it is important to explore adoptive therapies to supplement the host with a functional immune defense. In this study, we describe a virus carrier state that results from introducing lymphocytic choriomeningitis virus (LCMV) into adult mice possessing a restricted T cell repertoire. On infection of these mice, LCMV establishes systemic persistence, and within the CNS the virus infects astrocytes (and later oligodendrocytes) rather than its traditional parenchymal target neurons. To determine whether LCMV could be purged from a novel target selection in the absence of an endogenous immune repertoire, we adoptively transferred virus-specific memory cells into adult carrier mice. The memory cells purged virus from the periphery as well as the CNS, but they induced fatalities not typically associated with adoptive immunotherapy. When the repertoire of the recipient mice was examined, a deficiency in natural regulatory T cells was noted. We therefore supplemented carrier mice with regulatory T cells and simultaneously performed adoptive immunotherapy. Cotransfer of regulatory T cells significantly reduced mortality while still permitting the antiviral memory cells to purge the persistent infection. These data indicate that regulatory T cells can be used therapeutically to lessen the pathogenicity of virus-specific immune cells in an immunodeficient host. We also propose that the novel carrier state described herein will facilitate the study of immunotherapeutic regimens.
Collapse
Affiliation(s)
- Phi Truong
- Department of Immunology, Harold L Dorris Neurological Research Institute, Scripps Research Institute, La Jolla, CA 92037, USA
| | | |
Collapse
|
43
|
Kang SS, McGavern DB. Lymphocytic choriomeningitis infection of the central nervous system. FRONTIERS IN BIOSCIENCE : A JOURNAL AND VIRTUAL LIBRARY 2008; 13:4529-43. [PMID: 18508527 PMCID: PMC5279998 DOI: 10.2741/3021] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Viral infection of the central nervous system (CNS) can result in a multitude of responses including pathology, persistence or immune clearance. Lymphocytic choriomeningitis virus (LCMV) is a powerful model system to explore these potential outcomes of CNS infection due to the diversity of responses that can be achieved after viral inoculation. Several factors including tropism, timing, dose and variant of LCMV in combination with the development or suppression of the corresponding immune response dictates whether lethal meningitis, chronic infection or clearance of LCMV in the CNS will occur. Importantly, the functionality and positioning of the LCMV-specific CD8+ T cell response are critical in directing the subsequent outcome of CNS LCMV infection. Although a basic understanding of LCMV and immune interactions in the brain exists, the molecular machinery that shapes the balance between pathogenesis and clearance in the LCMV-infected CNS remains to be elucidated. This review covers the various outcomes of LCMV infection in the CNS and what is currently known about the impact of the virus itself versus the immune response in the development of disease or clearance.
Collapse
Affiliation(s)
- Silvia S. Kang
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, La Jolla, CA 92037
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037
| | - Dorian B. McGavern
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, La Jolla, CA 92037
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037
- Harold L. Dorris Neurological Research Institute, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
44
|
Recher M, Lang KS, Navarini A, Hunziker L, Lang PA, Fink K, Freigang S, Georgiev P, Hangartner L, Zellweger R, Bergthaler A, Hegazy AN, Eschli B, Theocharides A, Jeker LT, Merkler D, Odermatt B, Hersberger M, Hengartner H, Zinkernagel RM. Extralymphatic virus sanctuaries as a consequence of potent T-cell activation. Nat Med 2007; 13:1316-23. [PMID: 17982463 PMCID: PMC7096094 DOI: 10.1038/nm1670] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2007] [Accepted: 09/25/2007] [Indexed: 02/02/2023]
Abstract
T helper cells can support the functions of CD8(+) T cells against persistently infecting viruses such as murine lymphocytic choriomeningitis virus (LCMV), cytomegalovirus, hepatitis C virus and HIV. These viruses often resist complete elimination and remain detectable at sanctuary sites, such as the kidneys and other extralymphatic organs. The mechanisms underlying this persistence are not well understood. Here we show that mice with potent virus-specific T-cell responses have reduced levels and delayed formation of neutralizing antibodies, and these mice fail to clear LCMV from extralymphatic epithelia. Transfer of virus-specific B cells but not virus-specific T cells augmented virus clearance from persistent sites. Virus elimination from the kidneys was associated with the formation of IgG deposits in the interstitial space, presumably from kidney-infiltrating B cells. CD8(+) T cells in the kidneys of mice that did not clear virus from this site were activated but showed evidence of exhaustion. Thus, we conclude that in this model of infection, site-specific virus persistence develops as a consequence of potent immune activation coupled with reductions in virus-specific neutralizing antibodies. Our results suggest that sanctuary-site formation depends both on organ anatomy and on the induction of different adaptive immune effector mechanisms. Boosting T-cell responses alone may not reduce virus persistence.
Collapse
Affiliation(s)
- Mike Recher
- Institute for Experimental Immunology, University Hospital Zürich, Schmelzbergstrasse 12, CH-8091 Zürich, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Mothé BR, Stewart BS, Oseroff C, Bui HH, Stogiera S, Garcia Z, Dow C, Rodriguez-Carreno MP, Kotturi M, Pasquetto V, Botten J, Crotty S, Janssen E, Buchmeier MJ, Sette A. Chronic lymphocytic choriomeningitis virus infection actively down-regulates CD4+ T cell responses directed against a broad range of epitopes. THE JOURNAL OF IMMUNOLOGY 2007; 179:1058-67. [PMID: 17617598 DOI: 10.4049/jimmunol.179.2.1058] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Activation of CD4(+) T cells helps establish and sustain CD8(+) T cell responses and is required for the effective clearance of acute infection. CD4-deficient mice are unable to control persistent infection and CD4(+) T cells are usually defective in chronic and persistent infections. We investigated the question of how persistent infection impacted pre-existing lymphocytic choriomeningitis virus (LCMV)-specific CD4(+) T cell responses. We identified class II-restricted epitopes from the entire set of open reading frames from LCMV Armstrong in BALB/c mice (H-2(d)) acutely infected with LCMV Armstrong. Of nine epitopes identified, six were restricted by I-A(d), one by I-E(d) and two were dually restricted by both I-A(d) and I-E(d) molecules. Additional experiments revealed that CD4(+) T cell responses specific for these epitopes were not generated following infection with the immunosuppressive clone 13 strain of LCMV. Most importantly, in peptide-immunized mice, established CD4(+) T cell responses to these LCMV CD4 epitopes as well as nonviral, OVA-specific responses were actively suppressed following infection with LCMV clone 13 and were undetectable within 12 days after infection, suggesting an active inhibition of established helper responses. To address this dysfunction, we performed transfer experiments using both the Smarta and OT-II systems. OT-II cells were not detected after clone 13 infection, indicating physical deletion, while Smarta cells proliferated but were unable to produce IFN-gamma, suggesting impairment of the production of this cytokine. Thus, multiple mechanisms may be involved in the impairment of helper responses in the setting of early persistent infection.
Collapse
Affiliation(s)
- Bianca R Mothé
- Department of Biological Sciences, California State University, San Marcos, California 92096, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Koehn BH, Williams MA, Borom K, Gangappa S, Pearson TC, Ahmed R, Larsen CP. Fully MHC-Disparate Mixed Hemopoietic Chimeras Show Specific Defects in the Control of Chronic Viral Infections. THE JOURNAL OF IMMUNOLOGY 2007; 179:2616-26. [PMID: 17675525 DOI: 10.4049/jimmunol.179.4.2616] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The establishment of mixed allogeneic chimerism can induce donor-specific transplantation tolerance across full MHC barriers. However, a theoretical disadvantage of this approach is the possibility that the state of mixed chimerism might negatively affect the recipient's immune competence to control pathogens. Previous studies using murine models have not supported this hypothesis, because they indicate that acute viral infections are cleared by chimeric animals with similar kinetics to that of unmanipulated controls. However, chronic or persistent viral infections often require a more complex and sustained response with cooperation between CD4 Th cells, CTL, and B cells for effective control. The current study indicates that profound defects become manifest in the control of chronic pathogenic infections in MHC-disparate mixed allogeneic chimeric mice. Furthermore, we show that ineffective priming of the donor-restricted CTL response leads to virus persistence, as well as severe T cell exhaustion. Our results further suggest that either T cell adoptive immunotherapy or selected MHC haplotype matching partially restore immune competence. These approaches may facilitate the translation of mixed chimerism therapeutic regimens.
Collapse
Affiliation(s)
- Brent H Koehn
- Emory Transplant Center and Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Ismail N, Crossley EC, Stevenson HL, Walker DH. Relative importance of T-cell subsets in monocytotropic ehrlichiosis: a novel effector mechanism involved in Ehrlichia-induced immunopathology in murine ehrlichiosis. Infect Immun 2007; 75:4608-20. [PMID: 17562770 PMCID: PMC1951155 DOI: 10.1128/iai.00198-07] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Infection with gram-negative monocytotropic Ehrlichia strains results in a fatal toxic shock-like syndrome characterized by a decreased number of Ehrlichia-specific CD4(+) Th1 cells, the expansion of tumor necrosis factor alpha (TNF-alpha)-producing CD8(+) T cells, and the systemic overproduction of interleukin-10 (IL-10) and TNF-alpha. Here, we investigated the role of CD4(+) and CD8(+) T cells in immunity to Ehrlichia and the pathogenesis of fatal ehrlichiosis caused by infection with low- and high-dose (10(3) and 10(5) bacterial genomes/mouse, respectively) ehrlichial inocula. The CD4(+) T-cell-deficient mice showed exacerbated susceptibility to a lethal high- or low-dose infection and harbored higher bacterial numbers than did wild-type (WT) mice. Interestingly, the CD8(+) T-cell-deficient mice were resistant to a low dose but succumbed to a high dose of Ehrlichia. The absence of CD8(+) T cells abrogated TNF-alpha and IL-10 production, reduced tissue injury and bacterial burden, restored splenic CD4(+) T-cell numbers, and increased the frequency of Ehrlichia-specific CD4(+) Th1 cells in comparison to infected WT mice. Although fatal disease is perforin independent, our data suggested that perforin played a critical role in controlling bacterial burden and mediating liver injury. Similar to WT mice, mortality of infected perforin-deficient mice was associated with CD4(+) T-cell apoptosis and a high serum concentration of IL-10. Depletion of IL-10 restored the number of CD4(+) and CD8(+) T cells in infected WT mice. Our data demonstrate a novel mechanism of immunopathology in which CD8(+) T cells mediate Ehrlichia-induced toxic shock, which is associated with IL-10 overproduction and CD4(+) T-cell apoptosis.
Collapse
Affiliation(s)
- Nahed Ismail
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, 301 University Blvd., Galveston, TX 77555-0609, USA.
| | | | | | | |
Collapse
|
48
|
Lauterbach H, Truong P, McGavern DB. Clearance of an immunosuppressive virus from the CNS coincides with immune reanimation and diversification. Virol J 2007; 4:53. [PMID: 17553158 PMCID: PMC1899484 DOI: 10.1186/1743-422x-4-53] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2007] [Accepted: 06/06/2007] [Indexed: 12/13/2022] Open
Abstract
Once a virus infection establishes persistence in the central nervous system (CNS), it is especially difficult to eliminate from this specialized compartment. Therefore, it is of the utmost importance to fully understand scenarios during which a persisting virus is ultimately purged from the CNS by the adaptive immune system. Such a scenario can be found following infection of adult mice with an immunosuppressive variant of lymphocytic choriomeningitis virus (LCMV) referred to as clone 13. In this study we demonstrate that following intravenous inoculation, clone 13 rapidly infected peripheral tissues within one week, but more slowly inundated the entire brain parenchyma over the course of a month. During the establishment of persistence, we observed that genetically tagged LCMV-specific cytotoxic T lymphocytes (CTL) progressively lost function; however, the severity of this loss in the CNS was never as substantial as that observed in the periphery. One of the most impressive features of this model system is that the peripheral T cell response eventually regains functionality at ~60–80 days post-infection, and this was associated with a rapid decline in virus from the periphery. Coincident with this "reanimation phase" was a massive influx of CD4 T and B cells into the CNS and a dramatic reduction in viral distribution. In fact, olfactory bulb neurons served as the last refuge for the persisting virus, which was ultimately purged from the CNS within 200 days post-infection. These data indicate that a functionally revived immune response can prevail over a virus that establishes widespread presence both in the periphery and brain parenchyma, and that therapeutic enhancement of an existing response could serve as an effective means to thwart long term CNS persistence.
Collapse
Affiliation(s)
- Henning Lauterbach
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, 10550 North Torrey Pines Rd., La Jolla, CA 92037, USA
| | - Phi Truong
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, 10550 North Torrey Pines Rd., La Jolla, CA 92037, USA
| | - Dorian B McGavern
- Molecular and Integrative Neurosciences Department, The Scripps Research Institute, 10550 North Torrey Pines Rd., La Jolla, CA 92037, USA
- Harold L. Dorris Neurological Research Institute, The Scripps Research Institute, 10550 North Torrey Pines Rd., La Jolla, CA 92037, USA
| |
Collapse
|
49
|
Hinkula J. Clarification of how HIV-1 DNA and protein immunizations may be better used to obtain HIV-1-specific mucosal and systemic immunity. Expert Rev Vaccines 2007; 6:203-12. [PMID: 17408370 DOI: 10.1586/14760584.6.2.203] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
More focused research on a mucosal HIV-1 vaccine is needed urgently. An increasing amount of collected data, using heterologous multimodality prime-booster strategies, suggest that an efficient and protective HIV-1 vaccine must generate broad, long-lasting HIV-specific CD8(+) cytotoxic T-lymphocyte and neutralizing antibody responses. In the mucosa, these responses would be most effective if a preferential stimulus of HIV-1 neutralizing secretory immunoglobulin A and G were obtained. The attractive property of mucosal immunization is the obtained mucosal and systemic immunity, whereas systemic immunization induces a more limited immunity, predominantly in systemic sites. These objectives will require new vaccine regimens, such as multiclade HIV DNA and protein vaccines (nef, tat, gag and env expressed in DNA plasmids) delivered onto mucosal surfaces with needle-free delivery methods, such as nasal drop, as well as oral and rectal/vaginal delivery, and should merit clinical trials.
Collapse
Affiliation(s)
- Jorma Hinkula
- Department of Molecular Virology, Linkoping University, Linkoping, Sweden.
| |
Collapse
|
50
|
Schulze Zur Wiesch J, Lauer GM, Timm J, Kuntzen T, Neukamm M, Berical A, Jones AM, Nolan BE, Longworth SA, Kasprowicz V, McMahon C, Wurcel A, Lohse AW, Lewis-Ximenez LL, Chung RT, Kim AY, Allen TM, Walker BD. Immunologic evidence for lack of heterologous protection following resolution of HCV in patients with non-genotype 1 infection. Blood 2007; 110:1559-69. [PMID: 17475911 PMCID: PMC1975840 DOI: 10.1182/blood-2007-01-069583] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Chronic hepatitis C virus (HCV) infection is typically characterized by a lack of virus-specific CD4(+) T-cell-proliferative responses, but strong responses have been described in a subset of persons with persistent viremia. One possible explanation for these responses is that they were primed by an earlier resolved infection and do not recognize the current circulating virus. We defined all targeted epitopes using overlapping peptides corresponding to a genotype 1a strain in 44 patients chronically infected with different HCV genotypes (GT). Surprisingly, more HCV-specific CD4(+) T-cell responses were detected in patients with chronic non-GT1 infection compared with patients with chronic GT1 infection (P = .017). Notably, we found serologic evidence of a previous exposure to GT1 in 4 patients with non-GT1 infection, and these persons also demonstrated significantly more responses than non-GT1 patients in whom genotype and HCV serotype were identical (P < .001). Comparison of recognition of GT1-specific peptides to peptides representing autologous virus revealed the absence of cross-recognition of the autologous circulating virus. These data indicate that persistent HCV infection can occur in the presence of an HCV-specific T-cell response primed against a heterologous HCV strain, and suggest that clearance of 1 GT does not necessarily protect against subsequent exposure to a second GT.
Collapse
Affiliation(s)
- Julian Schulze Zur Wiesch
- Partners AIDS Research Center, Infectious Disease Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|