101
|
Racanelli V, Frassanito MA, Leone P, Brunetti C, Ruggieri S, Dammacco F. Bone marrow of persistently hepatitis C virus-infected individuals accumulates memory CD8+ T cells specific for current and historical viral antigens: a study in patients with benign hematological disorders. THE JOURNAL OF IMMUNOLOGY 2007; 179:5387-98. [PMID: 17911625 DOI: 10.4049/jimmunol.179.8.5387] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The role of virus-specific T cells in hepatitis C virus (HCV) pathogenesis is not clear. Existing knowledge on the frequency, phenotype, and behavior of these cells comes from analyses of blood and liver, but other lymphoid compartments that may be important sites for functionally mature T cells have not yet been analyzed. We studied HCV-specific T cells from bone marrow, in comparison to those from peripheral blood and liver biopsy tissue, from 20 persistently HCV-infected patients with benign hematological disorders. Bone marrow contained a sizeable pool of CD8(+) T cells specific for epitopes from structural and nonstructural HCV proteins. These cells displayed the same effector memory phenotype as liver-derived equivalents and the same proliferative potential as blood-derived equivalents but had greater antiviral effector functions such as Ag-specific cytotoxicity and IFN-gamma production. These features were not shared by influenza virus-specific CD8(+) T cells in the same bone marrow samples. Despite their highly differentiated phenotype and activated status, some bone marrow-resident HCV-specific CD8(+) T cells were not directed against the infecting virus but, instead, against historical HCV Ags (i.e., viral species of a previous infection or minor viral species of the current infection). These findings provide a snapshot view of the distribution, differentiation, and functioning of virus-specific memory T cells in patients with persistent HCV infection.
Collapse
Affiliation(s)
- Vito Racanelli
- Department of Internal Medicine and Clinical Oncology, University of Bari Medical School, Bari, Italy.
| | | | | | | | | | | |
Collapse
|
102
|
Koyama S, Ishii KJ, Kumar H, Tanimoto T, Coban C, Uematsu S, Kawai T, Akira S. Differential role of TLR- and RLR-signaling in the immune responses to influenza A virus infection and vaccination. THE JOURNAL OF IMMUNOLOGY 2007; 179:4711-20. [PMID: 17878370 DOI: 10.4049/jimmunol.179.7.4711] [Citation(s) in RCA: 238] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The innate immune system recognizes influenza A virus via TLR 7 or retinoic acid-inducible gene I in a cell-type specific manner in vitro, however, physiological function(s) of the MyD88- or interferon-beta promoter stimulator 1 (IPS-1)-dependent signaling pathways in antiviral responses in vivo remain unclear. In this study, we show that although either MyD88- or IPS-1-signaling pathway was sufficient to control initial antiviral responses to intranasal influenza A virus infection, mice lacking both pathways failed to show antiviral responses, resulting in increased viral load in the lung. By contrast, induction of B cells or CD4 T cells specific to the dominant hemagglutinin or nuclear protein Ags respectively, was strictly dependent on MyD88 signaling, but not IPS-1 signaling, whereas induction of nuclear protein Ag-specific CD8 T cells was not impaired in the absence of either MyD88 or IPS-1. Moreover, vaccination of TLR7- and MyD88-deficient mice with inactivated virus failed to confer protection against a lethal live virus challenge. These results strongly suggest that either the MyD88 or IPS-1 signaling pathway is sufficient for initial antiviral responses, whereas the protective adaptive immune responses to influenza A virus are governed by the TLR7-MyD88 pathway.
Collapse
Affiliation(s)
- Shohei Koyama
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Yamadaoka, Suita, Osaka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
103
|
Kaech SM, Wherry EJ. Heterogeneity and cell-fate decisions in effector and memory CD8+ T cell differentiation during viral infection. Immunity 2007; 27:393-405. [PMID: 17892848 PMCID: PMC3431921 DOI: 10.1016/j.immuni.2007.08.007] [Citation(s) in RCA: 473] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2007] [Indexed: 12/26/2022]
Abstract
Heterogeneity is a hallmark of the adaptive immune system. This is most evident in the enormous diversity of B and T cell antigen receptors. There is also heterogeneity within antiviral T cell populations, and subsets of effector and memory T cells now permeate our thinking about specialization of T cell responses to pathogens. It has been less clear, however, how heterogeneity in developing virus-specific effector and memory T cells is related to cell-fate decisions in the immune response, such as the generation long-lived memory T cells. Here we discuss recent findings that might help redefine how heterogeneity in antiviral T cell populations gives rise to T cell subsets with short- and long-lived cell fates.
Collapse
Affiliation(s)
- Susan M. Kaech
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06437, USA
| | - E. John Wherry
- Immunology Program, The Wistar Institute, Philadelphia, PA 19104, USA
| |
Collapse
|
104
|
Jenkins MR, Kedzierska K, Doherty PC, Turner SJ. Heterogeneity of effector phenotype for acute phase and memory influenza A virus-specific CTL. THE JOURNAL OF IMMUNOLOGY 2007; 179:64-70. [PMID: 17579022 DOI: 10.4049/jimmunol.179.1.64] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Ag-specific, CD8+ CTLs clear influenza A viruses from the lung via granzyme (Gzm) and perforin-dependent mechanisms. Ex vivo analysis of perforin-Gzm mRNA profiles demonstrated substantial heterogeneity in patterns of effector mRNA transcription of CD8+ D(b)NP(366)- or D(b)PA(224)-specific CTL. The only difference between the two epitope-specific sets was apparent very early after infection with similar molecular profiles seen in peak primary and secondary responses and in long-term memory. Surprisingly, memory T cells also expressed a diverse pattern of effector mRNA profile with an emphasis on GzmB and, surprisingly, GzmK. This analysis thus defines how naive, effector, and memory T cells differ in cytotoxic potential and provides novel insight into the molecular signatures of effector molecules observed at various stages after infection.
Collapse
Affiliation(s)
- Misty R Jenkins
- Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria 3010, Australia
| | | | | | | |
Collapse
|
105
|
Day EB, Zeng W, Doherty PC, Jackson DC, Kedzierska K, Turner SJ. The Context of Epitope Presentation Can Influence Functional Quality of Recalled Influenza A Virus-Specific Memory CD8+ T Cells. THE JOURNAL OF IMMUNOLOGY 2007; 179:2187-94. [PMID: 17675478 DOI: 10.4049/jimmunol.179.4.2187] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Lipopeptide constructs offer a novel strategy for eliciting effective cellular and humoral immunity by directly targeting the vaccine Ag to dendritic cells. Importantly, it is not known how closely immunity generated after lipopeptide vaccination mimics that generated after natural infection. We have used a novel lipopeptide vaccine strategy to analyze both the quantity and quality of CD8(+) T cell immunity to an influenza A virus epitope derived from the acidic polymerase protein (PA(224)) in B6 mice. Vaccination with the PA(224) lipopeptide resulted in accelerated viral clearance after subsequent influenza virus infection. The lipopeptide was also effective at recalling secondary D(b)PA(224) responses in the lung. Lipopeptide recalled D(b)PA(224)-specific CTL produced lower levels of IFN-gamma and TNF-alpha, but produced similar levels of IL-2 when compared with D(b)PA(224)-specific CTL recalled after virus infection. Furthermore, lipopeptide- and virus-recalled CTL demonstrated similar TCR avidity. Interestingly, lipopeptide administration resulted in expansion of D(b)PA(224)-specific CTL using a normally subdominant TCRBV gene segment. Overall, these results demonstrate that protective CTL responses elicited by lipopeptide vaccines can be correlated with TCR avidity, IL-2 production, and broad TCR repertoire diversity. Furthermore, factors that impact the quality of immunity are discussed. These factors are important considerations when evaluating the efficacy of novel vaccine strategies that target dendritic cells for eliciting cellular immunity.
Collapse
Affiliation(s)
- E Bridie Day
- Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
106
|
Keith MR, Levy RB. Transplant conditions determine the contribution of homeostatically expanded donor CD8 memory cells to host lymphoid reconstitution following syngeneic HCT. Exp Hematol 2007; 35:1303-15. [PMID: 17553613 DOI: 10.1016/j.exphem.2007.04.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2007] [Revised: 04/09/2007] [Accepted: 04/13/2007] [Indexed: 10/23/2022]
Abstract
OBJECTIVE To investigate the ability of donor CD8 memory (CD8 TM) cells to expand in recipients following syngeneic hematopoietic stem cell transplants (HCT). The influence of clinically important transplant parameters--conditioning level, delayed infusion and dose--on the homeostatic expansion and overall contribution of donor CD8 TM to host CD8 reconstitution was determined. MATERIALS AND METHODS Lymphopenia-induced CD8 TM homeostatic expansion was examined in a syngeneic murine HCT model. Antigen specific CD8 TM included both T-cell receptor transgenic and nontransgenic populations. An ex vivo technique using antigen, interleukin (IL)-2, and IL-15 was used to generate homogenous transgenic CD8 TM (i.e., central memory) and was adapted to enrich the heterogeneous nontransgenic CD8 population specific for a nonameric epitope. RESULTS Both transgenic and naturally occurring CD8 memory populations, derived in vivo or generated ex vivo, underwent a similar kinetic pattern of homeostatic expansion following transplantation into ablatively conditioned syngeneic recipients. Transplant parameters, i.e., lower conditioning, delayed infusion, and lower donor CD8 cell numbers shortened the period of expansion and lowered the steady-state numbers. CONCLUSIONS The pattern of CD8 TM expansion was dependent on conditioning levels, time of infusion, and dose. Transplantation of varying donor CD8 TM numbers demonstrated there was a maximal donor cell contribution to host CD8 reconstitution. The application of multiple well-defined memory CD8 populations supports the notion that these findings are characteristic of memory CD8 cells in general.
Collapse
|
107
|
Hikono H, Kohlmeier JE, Takamura S, Wittmer ST, Roberts AD, Woodland DL. Activation phenotype, rather than central- or effector-memory phenotype, predicts the recall efficacy of memory CD8+ T cells. ACTA ACUST UNITED AC 2007; 204:1625-36. [PMID: 17606632 PMCID: PMC2118640 DOI: 10.1084/jem.20070322] [Citation(s) in RCA: 253] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The contributions of different subsets of memory CD8+ T cells to recall responses at mucosal sites of infection are poorly understood. Here, we analyzed the CD8+ T cell recall responses to respiratory virus infection in mice and demonstrate that activation markers, such as CD27 and CD43, define three distinct subpopulations of memory CD8+ T cells that differ in their capacities to mount recall responses. These subpopulations are distinct from effector– and central–memory subsets, coordinately express other markers associated with activation status, including CXCR3, CD127, and killer cell lectin-like receptor G1, and are superior to CD62L in predicting the capacity of memory T cells to mediate recall responses. Furthermore, the capacity of vaccines to elicit these memory T cell subpopulations predicted the efficacy of the recall response. These findings extend our understanding of how recall responses are generated and suggest that activation and migration markers define distinct, and unrelated, characteristics of memory T cells.
Collapse
|
108
|
Turner SJ, Olivas E, Gutierrez A, Diaz G, Doherty PC. Disregulated influenza A virus-specific CD8+ T cell homeostasis in the absence of IFN-gamma signaling. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 178:7616-22. [PMID: 17548597 DOI: 10.4049/jimmunol.178.12.7616] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Recent studies indicate that IFN-gamma may influence both the expansion and the trafficking of virus-specific CD8+ CTL, though the effects are not necessarily consistent for different models of viral and bacterial disease. Influenza A virus infection of mice deficient for IFN-gamma (IFN-gamma(-/-)) or deficient for the IFN-gamma receptor 1 (IFNGR1(-/-)) was, when compared with the wild-type (WT) B6 controls, associated with increased Ag-specific CD8+ T cell counts in the spleen and mediastinal lymph nodes. At the same time, fewer of these CTL effectors were found in the bronchoalveolar lavage population recovered from the IFN-gamma(-/-) mice. Comparable effects were observed for WT mice treated with a neutralizing IFN-gamma-specific mAb. Transfer of WT memory Thy1.1(+) CD8+ populations into Thy1.2+ B6 IFN-gamma(-/-) or IFNGR1(-/-) mice followed by intranasal virus challenge demonstrated both that IFN-gamma produced by the host was important for the regulation of Ag-specific CTL numbers and that IFN-gamma was likely to act directly on the T cells themselves. In addition, the prevalence of CTLs undergoing apoptosis in spleen was lower when measured directly ex vivo for IFN-gamma(-/-) vs WT B6 mice. The present analysis is the first comprehensive demonstration that IFN-gamma signaling can differentially regulate both Ag-specific CTL homeostasis in secondary lymphoid organs and trafficking to a site of virus-induced pathology.
Collapse
Affiliation(s)
- Stephen J Turner
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | | | | | | | | |
Collapse
|
109
|
Abstract
Technological advances in recent years have allowed for an ever-expanding ability to analyze and quantify in vivo immune responses. MHC tetramers, intracellular cytokine staining, an increasing repertoire of transgenic and "knockout" mice, and the detailed characterization of a variety of infectious models have all facilitated more precise and definitive analyses of the generation and function of cytotoxic T lymphocytes (CTL). Understanding the mechanisms behind the differentiation of effector and memory CTL is of increasing importance to develop vaccination strategies against a variety of established and emerging infectious diseases. This review focuses on recent advances in our understanding of how effector and memory CTL differentiate and survive in vivo in response to viral or bacterial infection.
Collapse
Affiliation(s)
- Matthew A Williams
- Howard Hughes Medical Institute, Department of Immunology, University of Washington, Seattle, Washington 98195, USA.
| | | |
Collapse
|
110
|
Hahn CL, Liewehr FR. Innate Immune Responses of the Dental Pulp to Caries. J Endod 2007; 33:643-51. [PMID: 17509400 DOI: 10.1016/j.joen.2007.01.001] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2006] [Revised: 12/19/2006] [Accepted: 01/01/2007] [Indexed: 11/29/2022]
Abstract
Various cells and inflammatory mediators are involved in the initial pulpal responses to caries. This review focuses on the cellular, neuronal, and vascular components of pulpal innate responses to caries. Discussion will include dentinal fluid, odontoblasts, neuropeptides, and neurogenic inflammation, which are not classic immune components but actively participate in the inflammatory response as the caries progress pulpally. Summaries of innate immune cells as well as their cytokines and chemokines in healthy and reversible pulpitis tissues are presented.
Collapse
Affiliation(s)
- Chin-Lo Hahn
- Department of Endodontics, School of Dentistry, Virginia Commonwealth University, 520 North 12th Street, Richmond, Virginia 3298-0566, USA
| | | |
Collapse
|
111
|
Barbey C, Baumgaertner P, Devevre E, Rubio-Godoy V, Derre L, Bricard G, Guillaume P, Luescher IF, Liénard D, Cerottini JC, Romero P, Rufer N, Speiser DE. IL-12 controls cytotoxicity of a novel subset of self-antigen-specific human CD28+ cytolytic T cells. THE JOURNAL OF IMMUNOLOGY 2007; 178:3566-74. [PMID: 17339453 DOI: 10.4049/jimmunol.178.6.3566] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Activated CD8 T cells develop cytotoxicity against autologous cells bearing foreign Ags and self/tumor Ags. However, self-specific cytolysis needs to be kept under control to avoid overwhelming immunopathology. After peptide vaccination of melanoma patients, we studied molecular and functional properties of T cell subsets specific for the self/tumor Ag Melan-A/MART-1. Ex vivo analysis revealed three Ag-specific effector memory (EM) populations, as follows: CD28-negative EM (EM28(-)) T cells strongly expressing granzyme/perforin, and two EM28(+) subsets, one with high and the other with low level expression of these cytotoxic proteins. For further functional characterization, we generated 117 stable CD8 T cell clones by ex vivo flow cytometry-based sorting of these subsets. All EM28(-)-derived clones lysed target cells with high efficacy. In contrast, EM28(+)-derived clones were heterogenous, and could be classified in two groups, one with high and the other with low killing capacity, correlating with granzyme/perforin expression. High and low killer phenotypes remained surprisingly stable for several months. However, strongly increased granzyme expression and cytotoxicity were observed after exposure to IL-12. Thus, the data reveal a newly identified subset of CD28(+) conditional killer T cells. Because CD28 can mediate strong costimulatory signals, tight cytotoxicity control, as shown in this study through IL-12, may be particularly important for subsets of T cells expressing CD28.
Collapse
Affiliation(s)
- Catherine Barbey
- Division of Clinical Onco-Immunology, Ludwig Institute for Cancer Research, Centre Hospitalier Universitaire Vaudois, Avenue Pierre-Decker 4, CH-1005 Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
112
|
Kohlmeier JE, Miller SC, Woodland DL. Cutting edge: Antigen is not required for the activation and maintenance of virus-specific memory CD8+ T cells in the lung airways. THE JOURNAL OF IMMUNOLOGY 2007; 178:4721-5. [PMID: 17404250 DOI: 10.4049/jimmunol.178.8.4721] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Respiratory virus infections establish a population of memory CD8(+) T cells in the lung airways that persist for months after infection. However, the relationship between Ag-specific memory T cells in the lung airways and the systemic memory T cell pool is not well understood. The majority of lung airway memory T cells express a highly activated phenotype (CD69(+)/CD127(-)), suggesting that recent Ag stimulation is required to drive T cell activation and recruitment to the lung airways. In this study, we demonstrate that the lung airway environment itself in the absence of cognate Ag alters the expression of acute activation markers such as CD69 and CD127 on memory CD8(+) T cells. Furthermore, the steady-state recruitment of virus-specific memory CD8(+) T cells to the lung airways from the circulation can occur without recent Ag stimulation. These findings alter the current perceptions concerning the contribution of Ag to the maintenance of peripheral T cell memory.
Collapse
|
113
|
Garulli B, Meola M, Stillitano MG, Kawaoka Y, Castrucci MR. Efficient vagina-to-lower respiratory tract immune trafficking in a murine model of influenza A virus infection. Virology 2007; 361:274-82. [PMID: 17222437 DOI: 10.1016/j.virol.2006.12.001] [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] [Received: 07/21/2006] [Revised: 08/23/2006] [Accepted: 12/01/2006] [Indexed: 11/30/2022]
Abstract
Effective vaccination strategies for infectious diseases take into account the induction, long-term maintenance and recall of memory T-cell populations. To understand the immunological cross-talk within the mucosal compartments, we compared intranasal to vaginal immunization and demonstrated that vaginal infection of BALB/c mice with influenza A virus provides protective mucosal immunity against both homosubtypic and heterosubtypic virus challenge in the respiratory tract. We found that, prior to the viral challenge, in vaginally primed mice, antigen-specific CD8+ T cells were not detected in the lung airways and levels of serum antibodies were lower than those observed in intranasally immunized mice. However, following pulmonary challenge, NP147-specific CD8+ T cells were recruited and amplified in vaginally primed mice to the same extent as those in intranasally primed mice. Thus, the long-term memory immune response elicited by vaginal immunization with influenza virus is efficiently recalled and offers reasonable protection against infection in the respiratory tract.
Collapse
Affiliation(s)
- Bruno Garulli
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | | | | | | | | |
Collapse
|
114
|
Smith AG, Sheridan PA, Harp JB, Beck MA. Diet-induced obese mice have increased mortality and altered immune responses when infected with influenza virus. J Nutr 2007; 137:1236-43. [PMID: 17449587 DOI: 10.1093/jn/137.5.1236] [Citation(s) in RCA: 281] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Obesity is associated with an impaired immune response, an increased susceptibility to bacterial infection, and a chronic increase in proinflammatory cytokines such as IL-6 and TNFalpha. However, few studies have examined the effect of obesity on the immune response to viral infections. Because infection with influenza is a leading cause of morbidity and mortality worldwide, we investigated the effect of obesity on early immune responses to influenza virus exposure. Diet-induced obese and lean control C57BL/6 mice were infected with influenza A/PR8/34, and lung pathology and immune responses were examined at d 0 (uninfected), 3, and 6, postinfection. Following infection, diet-induced obese mice had a significantly higher mortality rate than the lean controls and elevated lung pathology. Antiviral and proinflammatory cytokine mRNA production in the lungs of the infected mice was markedly different between obese and lean mice. IFNalpha and beta were only minimally expressed in the infected lungs of obese mice and there was a notable delay in expression of the proinflammatory cytokines IL-6 and TNFalpha. Additionally, obese mice had a substantial reduction in NK cell cytotoxicity. These data indicate that obesity inhibits the ability of the immune system to appropriately respond to influenza infection and suggests that obesity may lead to increased morbidity and mortality from viral infections.
Collapse
Affiliation(s)
- Alexia G Smith
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | |
Collapse
|
115
|
Keating R, Yue W, Rutigliano JA, So J, Olivas E, Thomas PG, Doherty PC. Virus-specific CD8+ T cells in the liver: armed and ready to kill. THE JOURNAL OF IMMUNOLOGY 2007; 178:2737-45. [PMID: 17312116 DOI: 10.4049/jimmunol.178.5.2737] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Influenza A virus infection of C57BL/6 mice is a well-characterized model for studying CD8+ T cell-mediated immunity. Analysis of primary and secondary responses showed that the liver is highly enriched for CD8+ T cells specific for the immunodominant H2D(b)NP(366-374) (D(b)NP(366)) epitope. Functional analysis established that these liver-derived virus-specific CD8+ T cells are fully competent cytotoxic effectors and IFN-gamma secretors. In addition, flow cytometric analysis of early apoptotic cells showed that these influenza-specific CD8+ T cells from liver are as viable as those in the spleen, bronchoalveolar lavage, mediastinal lymph nodes, or lung. Moreover, cytokine profiles of the influenza-specific CD8+ T cells recovered from different sites were consistent with the bronchoalveolar lavage, rather than liver population, being the most susceptible to activation-induced cell death. Importantly, adoptively transferred influenza virus-specific CD8+ T cells from the liver survived and were readily recalled after virus challenge. Together, these results show clearly that the liver is not a "graveyard" for influenza virus-specific CD8+ T cells.
Collapse
Affiliation(s)
- Rachael Keating
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | | | | | | | | | | |
Collapse
|
116
|
Belz GT, Zhang L, Lay MDH, Kupresanin F, Davenport MP. Killer T cells regulate antigen presentation for early expansion of memory, but not naive, CD8+ T cell. Proc Natl Acad Sci U S A 2007; 104:6341-6. [PMID: 17400753 PMCID: PMC1840050 DOI: 10.1073/pnas.0609990104] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Antigen presentation within the lymph node draining a site of infection is crucial for initiation of cytotoxic T cell responses. Precisely how this antigen presentation regulates T cell expansion in vivo is unclear. Here, we show that, in primary infection, antigen presentation peaks approximately 3 days postinfection and then slowly decays until day 12. This prolonged antigen presentation is required for optimal expansion of naive CD8(+) T cells, because early ablation of dendritic cells reduces the later CD8(+) T cell response. Antigen presentation during secondary infection was 10-fold lower in magnitude and largely terminated by day 4 postinfection. Expansion of memory, but not naive, antigen-specific T cells was tightly controlled by perforin-dependent cytolysis of antigen-presenting cells. The ability of the memory T cells to remove antigen-presenting cells provides a negative-feedback loop to directly limit the duration of antigen presentation in vivo.
Collapse
Affiliation(s)
- Gabrielle T. Belz
- *Division of Immunology, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Melbourne, Victoria 3050, Australia; and
- To whom correspondence may be addressed. E-mail: or
| | - Lei Zhang
- Department of Haematology, Prince of Wales Hospital and Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia
| | - Matthew D. H. Lay
- Department of Haematology, Prince of Wales Hospital and Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia
| | - Fiona Kupresanin
- *Division of Immunology, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Melbourne, Victoria 3050, Australia; and
| | - Miles P. Davenport
- Department of Haematology, Prince of Wales Hospital and Centre for Vascular Research, University of New South Wales, Kensington, New South Wales 2052, Australia
- To whom correspondence may be addressed. E-mail: or
| |
Collapse
|
117
|
Denton AE, Doherty PC, Turner SJ, La Gruta NL. IL-18, but not IL-12, is required for optimal cytokine production by influenza virus-specific CD8+ T cells. Eur J Immunol 2007; 37:368-75. [PMID: 17219365 DOI: 10.1002/eji.200636766] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The potent innate cytokines IL-12 and IL-18 are considered to be important antigen-independent mediators of IFN-gamma production by NK cells and T lymphocytes. The present analysis addresses the physiological role of IL-12 and IL-18 in the generation of virus-specific CD8+ T cells. Both wt C57BL/6J (B6) mice and mice with disrupted IL-12p40 (IL-12p40(-/-)) or IL-18 (IL-18(-/-)) genes were infected with an influenza A virus and the characteristics of the resultant epitope-specific CD8+ T cell responses were compared. While IL-12 appeared to have no notable effect on either virus growth or on CD8+ T cell response profiles, the absence of IL-18 was associated with delayed virus clearance from the lung and, despite normal numbers, a significantly reduced production of IFN-gamma, TNF-alpha, and IL-2 by epitope-specific CD8+ T cells. While this cytokine phenotype was broadly maintained in IL-12p40/IL-18 double-knockout mice, no evidence was seen for any additive effect. Together, our results suggest that IL-18, but not IL-12, induces optimal, antigen-specific production of key cytokines by CD8+ T cells for the efficient clearance of influenza virus from the lungs of infected mice.
Collapse
Affiliation(s)
- Alice E Denton
- Department of Microbiology and Immunology, University of Melbourne, Parkville, Australia
| | | | | | | |
Collapse
|
118
|
Stambas J, Doherty PC, Turner SJ. An in vivo cytotoxicity threshold for influenza A virus-specific effector and memory CD8(+) T cells. THE JOURNAL OF IMMUNOLOGY 2007; 178:1285-92. [PMID: 17237374 DOI: 10.4049/jimmunol.178.3.1285] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Influenza A virus infection of C57BL/6 (B6) mice is characterized by prominent CD8(+) T cell responses to H2D(b) complexed with peptides from the viral nucleoprotein (NP(366), ASNENMETM) and acid polymerase (PA(224), SSLENFRAYV). An in vivo cytotoxicity assay that depends on the adoptive transfer of peptide-pulsed, syngeneic targets was used in this study to quantitate the cytotoxic potential of D(b)NP(366)- and D(b)PA(224)-specific acute and memory CD8(+) T cells following primary or secondary virus challenge. Both T cell populations displayed equivalent levels of in vivo effector function when comparable numbers were transferred into naive B6 hosts. Cytotoxic activity following primary infection clearly correlated with the frequency of tetramer-stained CD8(+) T cells. This relationship looked, however, to be less direct following secondary exposure, partly because the numbers of CD8(+)D(b)NP(366)(+) T cells were greatly in excess. However, calculating the in vivo E:T ratios indicated that in vivo lysis, like many other biological functions, is threshold dependent. Furthermore, the capacity to eliminate peptide-pulsed targets was independent of the differentiation state (i.e., primary or secondary effectors) and was comparable for the two T cell specificities that were analyzed. These experiments provide insights that may be of value for adoptive immunotherapy, where careful consideration of both the activation state and the number of effector cells is required.
Collapse
Affiliation(s)
- John Stambas
- Department of Microbiology and Immunology, University of Melbourne, Parkville, Melbourne, Australia
| | | | | |
Collapse
|
119
|
Murphy A, Westwood JA, Brown LE, Teng MWL, Moeller M, Xu Y, Smyth MJ, Hwu P, Darcy PK, Kershaw MH. Antitumor activity of dual-specific T cells and influenza virus. Cancer Gene Ther 2007; 14:499-508. [PMID: 17332777 DOI: 10.1038/sj.cgt.7701034] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Activation and expansion of T cells are important in disease resolution, but tumors do not usually satisfy these immune requirements. Therefore, we employed a novel strategy whereby dual-specific T cells were generated that could respond to both tumor and influenza virus, reasoning that immunization with influenza virus would activate and expand tumor-specific cells, and inhibit tumor growth. Dual-specific T cells were generated by gene modification of influenza virus-specific mouse T cells with a chimeric gene-encoding reactivity against the erbB2 tumor-associated antigen. Dual-specific T cells were demonstrated to respond against both tumor and influenza in vitro, and expanded in vitro in response to influenza to a much greater degree than in response to tumor cells. Following adoptive transfer and immunization of tumor-bearing mice with influenza virus, dual-specific T cells expanded greatly in numbers in the peritoneal cavity and spleen. This resulted in a significant increase in time of survival of mice. However, tumors were not eradicated, which may have been due to the observed poor penetration of tumor by T cells. This is the first demonstration that the potent immunogenic nature of an infectious agent can be utilized to directly impact on T-cell expansion and activity against tumor in vivo.
Collapse
Affiliation(s)
- A Murphy
- Cancer Immunology Research Program, Peter MacCallum Cancer Centre, Melbourne, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Agrewala JN, Brown DM, Lepak NM, Duso D, Huston G, Swain SL. Unique Ability of Activated CD4+ T Cells but Not Rested Effectors to Migrate to Non-lymphoid Sites in the Absence of Inflammation. J Biol Chem 2007; 282:6106-15. [PMID: 17197446 DOI: 10.1074/jbc.m608266200] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Recent studies suggest that effector T cells generated by immune responses migrate to multiple non-lymphoid sites, even those without apparent expression of antigen or inflammation. To investigate the ability of distinct CD4(+) T lymphocyte subsets to enter and persist in non-lymphoid, noninflamed compartments, we examined the migration and persistence of naïve, effector, and rested effector CD4(+) T cells generated in vitro following transfer to nonimmunized adoptive hosts. Th1 and Th2 effectors migrated to both lymphoid and non-lymphoid organs (peritoneum, fat pads, and lung). In contrast, rested effectors and naïve cells migrated only to lymphoid areas. Adhesion molecule expression, but not chemokine receptor expression, correlated with the ability to enter non-lymphoid sites. Donor cells persisted longer in lymphoid than in non-lymphoid sites. When hosts with naïve and memory donor cells were challenged with antigen, effectors developed in situ, which also migrated to non-lymphoid sites. Memory cells showed an accelerated shift to non-lymphoid migration, in keeping with memory effector formation. These results suggest that only recently activated effector T cells can disperse to non-lymphoid sites in the absence of antigen and inflammation, and as effectors return to rest, they lose this ability. These data also argue that memory cells in lymphoid sites are longer lived and not in equilibrium with those in non-lymphoid sites.
Collapse
|
121
|
Abstract
UNLABELLED Activated CD8+ T cells migrate to the liver at the end of an immune response and go through apoptosis there, but this mechanism is impaired in mice lacking Toll-like receptor-4. This allowed us to test the importance of liver trapping in an ongoing immune response. In the absence of Toll-like receptor-4, reduced liver accumulation was associated with an increase in the circulating CD8+ T cell pool, more long-lived memory T cells and increased CD8+ T cell memory responses. Using experimental orthotopic liver transplantation, we showed that the effect of Toll-like receptor-4 on the formation of the CD8+ T cell memory resides in the liver. CONCLUSION These studies reveal a new function for the liver, which is to regulate the magnitude of T cell memory responses through a Toll-like receptor-4-dependent mechanism.
Collapse
Affiliation(s)
- Beena John
- The David H Smith Center for Vaccine Biology and Immunology, The Aab Institute for Biomedical Research, University of Rochester, Rochester, NY 14642, USA.
| | | | | |
Collapse
|
122
|
Lee S, Miller SA, Wright DW, Rock MT, Crowe JE. Tissue-specific regulation of CD8+ T-lymphocyte immunodominance in respiratory syncytial virus infection. J Virol 2006; 81:2349-58. [PMID: 17182672 PMCID: PMC1865932 DOI: 10.1128/jvi.01910-06] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cytotoxic T lymphocytes (CTLs) are critical for control of respiratory syncytial virus (RSV) infection in humans and mice. To investigate cellular immune responses to infection, it is important to identify major histocompatibility complex (MHC) class I-restricted CTL epitopes. In this study, we identified a new RSV-specific, H-2K(d)-restricted subdominant epitope in the M2 protein, M2(127-135) (amino acids 127 to 135). This finding allowed us to study the frequency of T lymphocytes responding to two H-2K(d)-presented epitopes in the same protein following RSV infection by enzyme-linked immunospot (ELISPOT) and intracellular cytokine assays for both lymphoid and nonlymphoid tissues. For the subdominant epitope, we identified an optimal nine-amino-acid peptide, VYNTVISYI, which contained an H-2K(d) consensus sequence with Y at position 2 and I at position 9. In addition, an MHC class I stabilization assay using TAP-2-deficient RMA-S cells transfected with K(d) or L(d) indicated that the epitope was presented by K(d). The ratios of T lymphocytes during the peak CTL response to RSV infection that were specific for M2(82-90) (dominant) to T lymphocytes specific for M2(127-135) (subdominant) were approximately 3:1 in the spleen and 10:1 in the lung. These ratios were observed consistently in primary or secondary infection by the ELISPOT assay and in secondary infection by MHC/peptide tetramer staining. The number of antigen-specific T lymphocytes dropped in the 6 weeks after infection; however, the proportions of T lymphocytes specific for the immunodominant and subdominant epitopes were maintained to a remarkable degree in a tissue-specific manner. These studies will facilitate investigation of the regulation of immunodominance of RSV-specific CTL epitopes.
Collapse
Affiliation(s)
- Sujin Lee
- Department of Pediatrics, Vanderbilt University Medical Center, T-2220 Medical Center North, 1161 21st Avenue South, Nashville, TN 37232-2905, USA
| | | | | | | | | |
Collapse
|
123
|
Zaunders JJ, Dyer WB, Munier ML, Ip S, Liu J, Amyes E, Rawlinson W, De Rose R, Kent SJ, Sullivan JS, Cooper DA, Kelleher AD. CD127+CCR5+CD38+++ CD4+ Th1 effector cells are an early component of the primary immune response to vaccinia virus and precede development of interleukin-2+ memory CD4+ T cells. J Virol 2006; 80:10151-61. [PMID: 17005692 PMCID: PMC1617315 DOI: 10.1128/jvi.02670-05] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The stages of development of human antigen-specific CD4+ T cells responding to viral infection and their differentiation into long-term memory cells are not well understood. The inoculation of healthy adults with vaccinia virus presents an opportunity to study these events intensively. Between days 11 and 14 postinoculation, there was a peak of proliferating CCR5+CD38+++ CD4+ effector cells which contained the cytotoxic granule marker T-cell intracellular antigen 1 and included gamma interferon (IFN-gamma)-producing vaccinia virus-specific CD4+ T cells. The majority of these initial vaccinia virus-specific CD4+ T cells were CD127+ and produced interleukin-2 (IL-2) but not CTLA-4 in response to restimulation in vitro. Between days 14 and 21, there was a switch from IFN-gamma and IL-2 coexpression to IL-2 production only, coinciding with a resting phenotype and an increased in vitro proliferation response. The early CCR5+CD38+++ vaccinia virus-specific CD4+ T cells were similar to our previous observations of human immunodeficiency virus (HIV)-specific CD4+ T cells in primary HIV type 1 (HIV-1) infection, but the vaccinia virus-specific cells expressed much more CD127 and IL-2 than we previously found in their HIV-specific counterparts. The current study provides important information on the differentiation of IL-2+ vaccinia virus-specific memory cells, allowing further study of antiviral effector CD4+ T cells in healthy adults and their dysfunction in HIV-1 infection.
Collapse
Affiliation(s)
- John J Zaunders
- Centre for Immunology, St. Vincent's Hospital, Victoria Street, Darlinghurst, NSW 2010, Australia.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
124
|
Jenkins MR, Webby R, Doherty PC, Turner SJ. Addition of a Prominent Epitope Affects Influenza A Virus-Specific CD8+T Cell Immunodominance Hierarchies When Antigen Is Limiting. THE JOURNAL OF IMMUNOLOGY 2006; 177:2917-25. [PMID: 16920927 DOI: 10.4049/jimmunol.177.5.2917] [Citation(s) in RCA: 126] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A reverse genetics strategy was used to insert the OVA peptide (amino acid sequence SIINFEKL; OVA(257-264)) into the neuraminidase stalk of both the A/PR8 (H1N1) and A/HKx31 (H3N2) influenza A viruses. Initial characterization determined that K(b)OVA257 is presented on targets infected with PR8-OVA and HK-OVA without significantly altering D(b) nucleoprotein (NP)366 presentation. There were similar levels of K(b)OVA257- and D(b)NP366-specific CTL expansion following both primary and secondary intranasal challenge. Interestingly, while variable, the presence of the immunodominant K(b)OVA257-specific response resulted in diminished D(b) acidic polymerase224- and K(b) basic polymerase subunit 1(703)-, but not D(b)NP366-specific responses and didn't alter endogenous influenza A virus-specific immunodominance hierarchies. However, challenging PR8-OVA-primed mice with HK-OVA via the i.p. route, and thereby limiting Ag dose, led to a reduction in the magnitude of all the influenza A virus-specific responses measured. A similar reduction in CTL response to native epitopes was also seen following primary respiratory HK-OVA infection of mice that received substantial numbers of K(b)OVA257-specific TCR transgenic T cells. Thus, during the course of infection, the generation of individual virus-specific CTL responses is independently regulated. However, in cases in which Ag is limiting, or high precursor frequency, the presence of immunodominant CTL responses can impact on the magnitude of other specific populations. Therefore, depending on both the size of the T cell precursor pool and the mode of Ag presentation, the addition of a major epitope can diminish the size of endogenous, influenza-specific CD8+ T cell responses, although never to the point that these are totally compromised.
Collapse
Affiliation(s)
- Misty Rayna Jenkins
- Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia
| | | | | | | |
Collapse
|
125
|
Hikono H, Kohlmeier JE, Ely KH, Scott I, Roberts AD, Blackman MA, Woodland DL. T‐cell memory and recall responses to respiratory virus infections. Immunol Rev 2006; 211:119-32. [PMID: 16824122 DOI: 10.1111/j.0105-2896.2006.00385.x] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The respiratory tract is characterized by its large surface area and the close association of an extensive vasculature with the external environment. As such, the respiratory tract is a major portal of entry for many pathogens. The immune system is able to effectively control most pulmonary pathogens and establish immunological memory that is capable of mediating an accelerated and enhanced recall response to secondary pathogen challenge. A key component of the recall response in the lung involves the rapid response of antigen-specific memory CD8+ T cells. Recent studies have shown that memory CD8+ T cells are extremely heterogeneous in terms of phenotype, function, anatomical distribution, and longevity. However, we have little understanding of how the different subsets of memory cells actually contribute to the recall response, especially with respect to peripheral or mucosal sites, such as the lung. Since immunological memory is the cornerstone of vaccination, it is essential that we understand how different memory CD8+ T-cell subsets are initially generated, maintained over time, and contribute to recall responses. This review focuses on memory T cells that mediate recall responses to influenza and parainfluenza virus infections in the lung.
Collapse
|
126
|
Kedzierska K, Venturi V, Field K, Davenport MP, Turner SJ, Doherty PC. Early establishment of diverse T cell receptor profiles for influenza-specific CD8(+)CD62L(hi) memory T cells. Proc Natl Acad Sci U S A 2006; 103:9184-9. [PMID: 16754852 PMCID: PMC1482587 DOI: 10.1073/pnas.0603289103] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Single-cell analysis of endogenous, primary CD8(+) T cell responses to the influenza D(b)NP(366) and D(b)PA(224) epitopes indicates that prominent clonotypes bearing "public" or "shared" T cell receptors (TCRs) subset early into CD62L(hi) and CD62L(lo) populations. The CD62L(lo) effectors divide more and are rapidly eliminated during the contraction phase, whereas stable CD62L(hi) memory populations persist in the long-term. Reflecting the high frequency of small CD62L(hi) clones expressing "private" TCRs, the TCR diversity range per mouse is generally two times higher within the CD62L(hi)CD8(+)D(b)NP(366)(+) set (1.6 times higher for CD62L(hi)CD8(+)D(b)PA(224)(+)) from 8 to >180 days after antigen challenge. Memory CD8(+)CD62L(hi) T cell precursors thus segregate from the outset into populations expressing "best-fit" and "suboptimal" TCR characteristics, with this pattern being maintained stably thereafter. Hence, our analysis suggests that early establishment of influenza-specific memory within the CD8(+)CD62L(hi) subset preserves clonal diversity and prevents "overdominance" by a few public, or shared, clones.
Collapse
Affiliation(s)
- Katherine Kedzierska
- *Department of Microbiology and Immunology, University of Melbourne, Parkville 3010, Melbourne, Australia
| | - Vanessa Venturi
- Department of Haematology, Prince of Wales Hospital and Centre for Vascular Research, University of New South Wales, Kensington 2052, Sydney, Australia; and
| | - Kenneth Field
- *Department of Microbiology and Immunology, University of Melbourne, Parkville 3010, Melbourne, Australia
| | - Miles P. Davenport
- Department of Haematology, Prince of Wales Hospital and Centre for Vascular Research, University of New South Wales, Kensington 2052, Sydney, Australia; and
| | - Stephen J. Turner
- *Department of Microbiology and Immunology, University of Melbourne, Parkville 3010, Melbourne, Australia
| | - Peter C. Doherty
- *Department of Microbiology and Immunology, University of Melbourne, Parkville 3010, Melbourne, Australia
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105
| |
Collapse
|
127
|
Kedzierska K, La Gruta NL, Turner SJ, Doherty PC. Establishment and recall of CD8
+
T‐cell memory in a model of localized transient infection. Immunol Rev 2006; 211:133-45. [PMID: 16824123 DOI: 10.1111/j.0105-2896.2006.00386.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The influenza A virus model of localized, transient respiratory infection provides a well-defined experimental system for dissecting the induction and maintenance of CD8+ T-cell memory. This review focuses on quantitative and qualitative aspects of the prominent D(b)NP366- and D(b)PA224-specific CD8+ T-cell responses in virus-infected B6 mice. The different virus-specific effector and memory sets are compared by phenotypic [CD62L, interleukin-7 receptor-alpha (IL-7Ralpha), and IL-15Rbeta expression] and functional [interferon-gamma (IFN-gamma), tumor necrosis factor-alpha (TNF-alpha), and IL-2 production] analyses. Most clonotypes [defined by T-cell receptor (TCR) CDR3beta sequence] generated during the acute phase of infection survive into memory, with those expressing the more consensus 'canonical' TCRs being the major contributors to the recall response. The extent of clonal expansion and the size of memory CD8+ T-cell populations has been characterized for mice challenged with either wildtype or mutant viruses, where broadly equivalent D(b)NP366 and D(b)PA224 expression was achieved by disabling the peptides in their native configuration, then expressing them in the viral neuraminidase protein. Combining the clonotypic and antigen dose analyses led to a somewhat mechanistic conclusion that the magnitude of any virus-specific CD8+ T-cell response will be a direct function of antigen dose and the size of the naïve or memory CD8+ T-cell precursor pool.
Collapse
Affiliation(s)
- Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, Parkville, Melbourne, Australia
| | | | | | | |
Collapse
|
128
|
Kohlmeier JE, Woodland DL. Memory T cell recruitment to the lung airways. Curr Opin Immunol 2006; 18:357-62. [PMID: 16616475 DOI: 10.1016/j.coi.2006.03.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2006] [Accepted: 03/28/2006] [Indexed: 11/15/2022]
Abstract
Memory T cells play an important role in the resolution of secondary respiratory virus infections through the production of anti-viral cytokines and the lysis of infected cells. Within the lung airways, memory T cells form an initial line of defense against secondary virus challenge by limiting early viral replication at the site of infection. Recent studies have addressed the generation and maintenance of airway memory T cells and their relationship with memory T cells in the secondary lymphoid organs. These advances have demonstrated that different memory subsets contribute to the maintenance of the airway memory T cell population and have identified molecules that influence trafficking to the lung tissue and the lung airways.
Collapse
Affiliation(s)
- Jacob E Kohlmeier
- Trudeau Institute, 154 Algonquin Avenue, Saranac Lake, New York, NY 12983, USA
| | | |
Collapse
|
129
|
Swain SL, Agrewala JN, Brown DM, Jelley-Gibbs DM, Golech S, Huston G, Jones SC, Kamperschroer C, Lee WH, McKinstry KK, Román E, Strutt T, Weng NP. CD4+ T-cell memory: generation and multi-faceted roles for CD4+ T cells in protective immunity to influenza. Immunol Rev 2006; 211:8-22. [PMID: 16824113 PMCID: PMC2266984 DOI: 10.1111/j.0105-2896.2006.00388.x] [Citation(s) in RCA: 148] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We have outlined the carefully orchestrated process of CD4+ T-cell differentiation from naïve to effector and from effector to memory cells with a focus on how these processes can be studied in vivo in responses to pathogen infection. We emphasize that the regulatory factors that determine the quality and quantity of the effector and memory cells generated include (i) the antigen dose during the initial T-cell interaction with antigen-presenting cells; (ii) the dose and duration of repeated interactions; and (iii) the milieu of inflammatory and growth cytokines that responding CD4+ T cells encounter. We suggest that heterogeneity in these regulatory factors leads to the generation of a spectrum of effectors with different functional attributes. Furthermore, we suggest that it is the presence of effectors at different stages along a pathway of progressive linear differentiation that leads to a related spectrum of memory cells. Our studies particularly highlight the multifaceted roles of CD4+ effector and memory T cells in protective responses to influenza infection and support the concept that efficient priming of CD4+ T cells that react to shared influenza proteins could contribute greatly to vaccine strategies for influenza.
Collapse
|
130
|
Selin LK, Brehm MA, Naumov YN, Cornberg M, Kim SK, Clute SC, Welsh RM. Memory of mice and men: CD8+ T-cell cross-reactivity and heterologous immunity. Immunol Rev 2006; 211:164-81. [PMID: 16824126 PMCID: PMC7165519 DOI: 10.1111/j.0105-2896.2006.00394.x] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The main functions of memory T cells are to provide protection upon re-exposure to a pathogen and to prevent the re-emergence of low-grade persistent pathogens. Memory T cells achieve these functions through their high frequency and elevated activation state, which lead to rapid responses upon antigenic challenge. The significance and characteristics of memory CD8+ T cells in viral infections have been studied extensively. In many of these studies of T-cell memory, experimental viral immunologists go to great lengths to assure that their animal colonies are free of endogenous pathogens in order to design reproducible experiments. These experimental results are then thought to provide the basis for our understanding of human immune responses to viruses. Although these findings can be enlightening, humans are not immunologically naïve, and they often have memory T-cell populations that can cross-react with and respond to a new infectious agent or cross-react with allo-antigens and influence the success of tissue transplantation. These cross-reactive T cells can become activated and modulate the immune response and outcome of subsequent heterologous infections, a phenomenon we have termed heterologous immunity. These large memory populations are also accommodated into a finite immune system, requiring that the host makes room for each new population of memory cell. It appears that memory cells are part of a continually evolving interactive network, where with each new infection there is an alteration in the frequencies, distributions, and activities of memory cells generated in response to previous infections and allo-antigens.
Collapse
Affiliation(s)
- Liisa K Selin
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | | | | | | | | | | | | |
Collapse
|
131
|
Abstract
The respiratory tract poses a substantial challenge to the immune system due to its large surface area, an extensive vasculature that is in very close proximity to the external environment, and repeated exposure to potentially pathogenic organisms in the air. Yet many lung pathogens are controlled by appropriate immune responses. The underlying mechanisms of the adaptive cellular immune response in protecting the respiratory tract are poorly understood. Recently, it has emerged that memory CD4(+) and CD8(+) T cells are present in the lung airways, and evidence is mounting that these cells play a key role in pulmonary immunity to pathogen challenge by immediately engaging the pathogen at the site of infection when pathogen loads are low. For example, in the case of respiratory virus infections, there is evidence that both CD4(+) and CD8(+) memory cells in the lung airways mediate substantial control of a secondary respiratory virus infection in the lungs. Here we address recent developments in our understanding of lung airway memory T cells and their role in infectious disease.
Collapse
Affiliation(s)
- David L Woodland
- Trudeau Institute, 154 Algonquin Avenue, Saranac Lake, NY 12983, USA.
| | | |
Collapse
|
132
|
Whitmire JK, Benning N, Whitton JL. Precursor frequency, nonlinear proliferation, and functional maturation of virus-specific CD4+ T cells. THE JOURNAL OF IMMUNOLOGY 2006; 176:3028-36. [PMID: 16493061 DOI: 10.4049/jimmunol.176.5.3028] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The early events regulating antiviral CD4 responses were tracked using an adoptive transfer model. CD4+ T cell expansion was nonlinear, with a lengthy lag phase followed by 2 days of explosive proliferation. A small number of naive Ag-specific CD4+ T cells were found in nonlymphoid tissues and, in the 8 days following infection, the number of activated cells increased in all tissues analyzed, and their effector functions matured. Finally, we show that a naive mouse contains approximately 100 naive CD4+ precursor cells specific for a single epitope, a precursor frequency of approximately 10(-5), similar to that of naive CD8+ T cells, indicating that the approximately 50-fold difference in size of the two responses to virus infection is determined by something other than the number of precursor cells.
Collapse
Affiliation(s)
- Jason K Whitmire
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
133
|
Abstract
Current vaccine strategies against influenza focus on generating robust antibody responses. Because of the high degree of antigenic drift among circulating influenza strains over the course of a year, vaccine strains must be reformulated specifically for each influenza season. The time delay from isolating the pandemic strain to large-scale vaccine production would be detrimental in a pandemic situation. A vaccine approach based on cell-mediated immunity that avoids some of these drawbacks is discussed here. Specifically, cell-mediated responses typically focus on peptides from internal influenza proteins, which are far less susceptible to antigenic variation. We review the literature on the role of CD4+ and CD8+ T cell-mediated immunity in influenza infection and the available data on the role of these responses in protection from highly pathogenic influenza infection. We discuss the advantages of developing a vaccine based on cell-mediated immune responses toward highly pathogenic influenza virus and potential problems arising from immune pressure.
Collapse
Affiliation(s)
- Paul G Thomas
- St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | | | | | | |
Collapse
|
134
|
Doherty PC, Turner SJ, Webby RG, Thomas PG. Influenza and the challenge for immunology. Nat Immunol 2006; 7:449-55. [PMID: 16622432 DOI: 10.1038/ni1343] [Citation(s) in RCA: 275] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2006] [Accepted: 02/22/2006] [Indexed: 01/15/2023]
Abstract
The continued westward dissemination of H5N1 influenza A viruses in avian populations and the nearly 50% mortality rate of humans infected with H5N1 are a source of great international concern. A mutant H5N1 virus with the capability to spread rapidly between humans could cause a global catastrophe. Governments have reacted by developing national response plans, stockpiling antiviral drugs and speeding up the development and approval of vaccines. Here we summarize what is known about the interaction between influenza A viruses and the mammalian host response, specifically emphasizing issues that might be of interest to the broader immunology community.
Collapse
Affiliation(s)
- Peter C Doherty
- Department of Microbiology and Immunology, University of Melbourne School of Medicine, Victoria 3010, Australia.
| | | | | | | |
Collapse
|
135
|
Deliyannis G, Kedzierska K, Lau YF, Zeng W, Turner SJ, Jackson DC, Brown LE. Intranasal lipopeptide primes lung-resident memory CD8+ T cells for long-term pulmonary protection against influenza. Eur J Immunol 2006; 36:770-8. [PMID: 16435281 DOI: 10.1002/eji.200535217] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The longevity of the influenza virus-specific CD8+ T cell response following intranasal delivery of a synthetic lipopeptide was investigated and the characteristics and location of the cells associated with viral clearance examined. The lipopeptide, incorporating an epitope for CD8+ T cells and another for CD4+ T cells with the lipid moiety S-[2,3-bis(palmitoyloxy)propyl]cysteine (Pam2Cys) attached, induced potent and long-lived pulmonary protection. Both the lipopeptide and its largely unprotective non-lipidated counterpart elicited comparable numbers of CD8+ T cells in the spleen, which was the main location of the memory pool. However, the lipopeptide, unlike the non-lipidated peptide, also induced a substantial memory population that remained in the lungs and was rapidly activated upon viral challenge months later. These lipopeptide-induced lung-resident CD8+ T cells were also very similar in number and IFN-gamma-secreting potential to those induced by prior exposure to the virus itself and are likely mediators of initial viral clearance prior to recruitment from the expanding lymph node T cell pool. Significant clearing responses were demonstrated as late as 9 months post-lipopeptide vaccination. This study shows that CD8+ T cells primed by the lipopeptide are not only long-lived but can take up residence in the lung where they are important early mediators of pulmonary protection.
Collapse
Affiliation(s)
- Georgia Deliyannis
- The Department of Microbiology and Immunology, The University of Melbourne, Parkville, Australia
| | | | | | | | | | | | | |
Collapse
|
136
|
Zammit DJ, Turner DL, Klonowski KD, Lefrançois L, Cauley LS. Residual antigen presentation after influenza virus infection affects CD8 T cell activation and migration. Immunity 2006; 24:439-49. [PMID: 16618602 PMCID: PMC2861289 DOI: 10.1016/j.immuni.2006.01.015] [Citation(s) in RCA: 218] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2005] [Revised: 01/25/2006] [Accepted: 01/31/2006] [Indexed: 11/25/2022]
Abstract
Activated virus-specific CD8 T cells remain in the lung airways for several months after influenza virus infection. We show that maintenance of this cell population is dependent upon the route of infection and prolonged presentation of viral antigen in the draining lymph nodes (DLN) of the respiratory tract. The local effects on T cell migration have been examined. We show retention of virus-specific CD8 T cells in the mediastinal lymph node (MLN) and continuing recruitment of blood-borne migrants into the lung airways during antigen presentation. These data show that antigen that is retained after pulmonary influenza virus infection controls the migratory pattern and activation state of virus-specific CD8 T cells near the site of virus amplification.
Collapse
Affiliation(s)
- David J. Zammit
- Department of Immunology University of Connecticut Health Center Farmington, Connecticut 06032
| | - Damian L. Turner
- Department of Immunology University of Connecticut Health Center Farmington, Connecticut 06032
| | - Kimberly D. Klonowski
- Department of Immunology University of Connecticut Health Center Farmington, Connecticut 06032
| | - Leo Lefrançois
- Department of Immunology University of Connecticut Health Center Farmington, Connecticut 06032
| | - Linda S. Cauley
- Department of Immunology University of Connecticut Health Center Farmington, Connecticut 06032
| |
Collapse
|
137
|
Hafalla JCR, Cockburn IA, Zavala F. Protective and pathogenic roles of CD8+ T cells during malaria infection. Parasite Immunol 2006; 28:15-24. [PMID: 16438672 DOI: 10.1111/j.1365-3024.2006.00777.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
CD8+ T cells play a key role in protection against pre-erythrocytic stages of malaria infection. Many vaccine strategies are based on the idea of inducing a strong infection-blocking CD8+ T cell response. Here, we summarize what is known about the development, specificity and protective effect of malaria-specific CD8+ T cells and report on recent developments in the field. Although work in mouse models continues to make progress in our understanding of the basic biology of these cells, many questions remain to be answered - particularly on the roles of these cells in human infections. Increasing evidence is also emerging of a harmful role for CD8+ T cells in the pathology of cerebral malaria in rodent systems. Once again, the relevance of these results to human disease is one of the primary questions facing workers in this field.
Collapse
Affiliation(s)
- J C R Hafalla
- Department of Medical Parasitology, New York University School of Medicine, New York, NY, USA
| | | | | |
Collapse
|
138
|
Thomas PG, Brown SA, Yue W, So J, Webby RJ, Doherty PC. An unexpected antibody response to an engineered influenza virus modifies CD8+ T cell responses. Proc Natl Acad Sci U S A 2006; 103:2764-9. [PMID: 16473934 PMCID: PMC1413843 DOI: 10.1073/pnas.0511185103] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The ovalbumin(323-339) peptide that binds H2I-A(b) was engineered into the globular heads of hemagglutinin (H) molecules from serologically non-cross-reactive H1N1 and H3N2 influenza A viruses, the aim being to analyze recall CD4+ T cell responses in a virus-induced respiratory disease. Prime/challenge experiments with these H1ova and H3ova viruses in H2(b) mice gave the predicted, ovalbumin-specific CD4+ T cell response but showed an unexpectedly enhanced, early expansion of viral epitope-specific CD8+ T cells in spleen and a greatly diminished inflammatory process in the virus-infected respiratory tract. At the same time, the primary antibody response to the H3N2 challenge virus was significantly reduced, an effect that has been associated with preexisting neutralizing antibody in other experimental systems. Analysis of serum from the H1ova-primed mice showed low-level binding to H3ova but not to the wild-type H3N2 virus. Experiments with CD4+ T cell-depleted and Ig-/- mice indicated that this cross-reactive Ig is indeed responsible for the modified pathogenesis after respiratory challenge. Furthermore, the effect does not seem to be virus-dose related, although it does require infection. These findings suggest intriguing possibilities for vaccination and, at the same time, emphasize that engineered modifications in viruses may have unintended immunological consequences.
Collapse
Affiliation(s)
| | | | - Wen Yue
- Departments of *Immunology and
| | | | - Richard J. Webby
- Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Peter C. Doherty
- Departments of *Immunology and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
139
|
Dare R, Sykes PJ, Morley AA, Brisco MJ. Effect of age on the repertoire of cytotoxic memory (CD8+CD45RO+) T cells in peripheral blood: the use of rearranged T cell receptor gamma genes as clonal markers. J Immunol Methods 2006; 308:1-12. [PMID: 16325196 DOI: 10.1016/j.jim.2005.08.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2005] [Revised: 08/23/2005] [Accepted: 08/31/2005] [Indexed: 11/24/2022]
Abstract
We have established a method to estimate the number of clones in peripheral blood, using rearranged T cell receptor gamma genes as clonal markers, selecting cells at random, and establishing the sizes of the clones to which they belong. Clone sizes were quantified by a clone-specific PCR test based on the VNJ junctional sequence, which typically detects 1-2 copies of its target gene. All clones chosen for study were subsequently quantified in blood, and sizes ranged from 3 x 10(-6) (1 cell in 330,000 CD8+CD45RO+ cells) to 3.5 x 10(-2) permitting numbers of clones to be estimated from the harmonic mean of clone size. Two independent estimates from a healthy young adult (20-30 years old) gave repertoires of 94,000 and 110,000 clones. Two other healthy young adults gave repertoires of 40,000 and 55,000 clones. Repertoires in four healthy active older (>75 years old) adults were more variable but generally lower, being 3600, 5500, 14,000 and 97,000 clones, despite enlarged clones making up >1% of the compartment in the last individual. Overall, young adults had smaller clones (p=0.026, non-directional Mann-Whitney U-test). If the human body contains 5 l of blood, clones have 2 x 10(3)-1.0 x 10(7) cells in blood. These results confirm a diverse repertoire of rearranged T cell receptor gamma genes. The number of clones thus defined are broadly consistent with other estimates of repertoire, despite differences in marker genes used and subsets of cells studied.
Collapse
Affiliation(s)
- Raellene Dare
- Department of Haematology and Genetic Pathology, Flinders University and Flinders Medical Centre, Bedford Park, South Australia 5042, Australia
| | | | | | | |
Collapse
|
140
|
Fadel SA, Cowell LG, Cao S, Ozaki DA, Kepler TB, Steeber DA, Sarzotti M. Neonate-primed CD8+ memory cells rival adult-primed memory cells in antigen-driven expansion and anti-viral protection. Int Immunol 2006; 18:249-57. [PMID: 16418189 DOI: 10.1093/intimm/dxh360] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Immunizations early in life, when the host is most susceptible to infection, allow protective immunological memory to develop. Decreasing the dose of Cas-Br-E murine leukemia virus when priming neonatal mice results in adult-like, Type 1 protective responses, but the resulting memory cell populations are smaller than after adult priming. After secondary challenge, virus-specific CD8+ memory cell populations expand twice as much in neonate-primed mice as in adult-primed mice. We found that when equivalent numbers of virus-specific cells were transferred into virus-susceptible mice, protection from disease was similar whether donor, immune mice were primed as neonates or adults, and IL-4 did not alter in vivo virus-specific CD8+ memory cell effector function. Hence, neonate-primed CD8+ cells develop into memory cells that rival adult-primed cells in proliferation and effector function.
Collapse
Affiliation(s)
- Shaza A Fadel
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | |
Collapse
|
141
|
Abstract
T cell responses to viral infections can mediate either protective immunity or damaging immunopathology. Viral infections induce the proliferation of T cells specific for viral antigens and cause a loss in the number of T cells with other specificities. In immunologically naive hosts, viruses will induce T cell responses that, dependent on the MHC, recognize a distinct hierarchy of virus-encoded T cell epitopes. This hierarchy can change if the host has previously encountered another pathogen that elicited a memory pool ofT cells specific to a cross-reactive epitope. This heterologous immunity can deviate the normal immune response and result in either beneficial or harmful effects on the host. Each host has a unique T cell repertoire caused by the random DNA rearrangement that created it, so the specific T cells that create the epitope hierarchy differ between individuals. This "private specificity" seems of little significance in the T cell response of a naive host to infection, but it is of profound importance under conditions of heterologous immunity, where a small subset of a cross-reactive memory pool may expand and dominate a response. Examples are given of how the private specificities of immune responses under conditions of heterologous immunity influence the pathogenesis of murine and human viral infections.
Collapse
Affiliation(s)
- Bali Pulendran
- Emory Vaccine Center, 954 Gatewood Road, Atlante, GA 30329 USA
| | - Rafi Ahmed
- Emory Vaccine Center, 954 Gatewood Road, Atlante, GA 30329 USA
| |
Collapse
|
142
|
Bachmann MF, Wolint P, Schwarz K, Oxenius A. Recall proliferation potential of memory CD8+ T cells and antiviral protection. THE JOURNAL OF IMMUNOLOGY 2005; 175:4677-85. [PMID: 16177115 DOI: 10.4049/jimmunol.175.7.4677] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Memory CD8+ T cells play a crucial role in mediating protection from infection with viruses and other intracellular pathogens. Memory T cells are not a homogenous cellular population and may be separated into central memory T cells with substantial recall proliferation capacity and effector memory T cells with limited recall proliferation capacity. It has been suggested that the protective capacity of effector memory T cells is more limited than that of central memory T cells in viral infections. Here, we show that pronounced recall proliferation potential is indeed key for protection against lymphocytic choriomeningitis virus, which replicates in central lymphoid organs and is controlled by contact-dependent lysis of infected cells. In contrast, recall proliferation competence is not sufficient for protection against vaccinia virus, which is replicating in peripheral solid organs and is controlled by cytokines. To protect against vaccinia virus, high numbers of effector-like T cells were required to be present in peripheral tissue before viral challenge. These data indicate that the protective capacity of different subpopulations of memory T cells may vary dependent on the nature and the route of the challenge infection, which must be considered in T cell-based vaccine design.
Collapse
|
143
|
Parretta E, Cassese G, Barba P, Santoni A, Guardiola J, Di Rosa F. CD8 cell division maintaining cytotoxic memory occurs predominantly in the bone marrow. THE JOURNAL OF IMMUNOLOGY 2005; 174:7654-64. [PMID: 15944266 DOI: 10.4049/jimmunol.174.12.7654] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Long-term persistence of Ag-experienced CD8 cells, a class of T lymphocytes with cytotoxic function, contributes to immunological memory against intracellular pathogens. After Ag clearance, memory CD8 cells are maintained over time by a slow proliferation, primarily cytokine driven. In this article, we show that the bone marrow (BM) is the crucial organ where such basal division of memory CD8 cells occurs. BM memory CD8 cells contain a higher percentage of proliferating cells than their corresponding cells in either spleen or lymph nodes from C57BL/6 mice. This occurs both in the case of memory-phenotype CD44(high) CD8 cells and in the case of Ag-specific memory CD8 cells. Importantly, the absolute number of Ag-specific memory CD8 cells dividing in the BM largely exceeds that in spleen, lymph nodes, liver, and lung taken together. In the BM, Ag-specific memory CD8 cells express lower levels of CD127, i.e., the alpha-chain of IL-7R, than in either spleen or lymph nodes. We interpret these results as indirect evidence that Ag-specific memory CD8 cells receive proliferative signals by IL-7 and/or IL-15 in the BM and propose that the BM acts as a saturable "niche" for the Ag-independent proliferation of memory CD8 cells. Taken together, our novel findings indicate that the BM plays a relevant role in the maintenance of cytotoxic T cell memory, in addition to its previously described involvement in long-term Ab responses.
Collapse
Affiliation(s)
- Elisabetta Parretta
- Institute of Genetics and Biophysics, Adriano Buzzati Traverso, Consiglio Nazionale delle Ricerche, Naples, Italy
| | | | | | | | | | | |
Collapse
|
144
|
Pan CH, Valsamakis A, Colella T, Nair N, Adams RJ, Polack FP, Greer CE, Perri S, Polo JM, Griffin DE. Modulation of disease, T cell responses, and measles virus clearance in monkeys vaccinated with H-encoding alphavirus replicon particles. Proc Natl Acad Sci U S A 2005; 102:11581-8. [PMID: 16037211 PMCID: PMC1187989 DOI: 10.1073/pnas.0504592102] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Measles remains a major worldwide problem partly because of difficulties with vaccination of young infants. New vaccine strategies need to be safe and to provide sustained protective immunity. We have developed Sindbis virus replicon particles that express the measles virus (MV) hemagglutinin (SIN-H) or fusion (SIN-F) proteins. In mice, SIN-H induced high-titered, dose-dependent, MV-neutralizing antibody after a single vaccination. SIN-F, or SIN-H and SIN-F combined, induced somewhat lower responses. To assess protective efficacy, juvenile macaques were vaccinated with a single dose of 10(6) or 10(8) SIN-H particles and infant macaques with two doses of 10(8) particles. A dose of 10(8) particles induced sustained levels of high-titered, MV-neutralizing antibody and IFN-gamma-producing memory T cells, and most monkeys were protected from rash when challenged with wild-type MV 18 months later. After challenge, there was a biphasic appearance of H- and F-specific IFN-gamma-secreting CD4+ and CD8+ T cells in vaccinated monkeys, with peaks approximately 1 and 3-4 months after challenge. Viremia was cleared within 14 days, but MV RNA was detectable for 4-5 months. These studies suggest that complete clearance of MV after infection is a prolonged, phased, and complex process influenced by prior vaccination.
Collapse
Affiliation(s)
- Chien-Hsiung Pan
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
145
|
Abstract
The ability of memory CD8 T cells to patrol non-lymphoid tissues represents an effective method whereby proficient immunosurveillance is achieved. From the analysis of memory CD8 T cell migration in vivo, it is clear that tissue-specific factors control trafficking and residence time within tissues. We propose that at least three pools of memory CD8 T cells exist based on migratory capabilities as dictated by their location in the body. Moreover, we hypothesize that the process of acquisition of homeostatic signals in specific tissues, such as the cytokines IL-7 and IL-15, regulates the mobility of memory T cells.
Collapse
Affiliation(s)
- Kimberly D Klonowski
- Division of Immunology, Department of Medicine, University of Connecticut Health Center, M/C 1319, 263 Farmington Avenue, Farmington, CT 06030-1319, USA
| | | |
Collapse
|
146
|
Kedzierska K, La Gruta NL, Davenport MP, Turner SJ, Doherty PC. Contribution of T cell receptor affinity to overall avidity for virus-specific CD8+ T cell responses. Proc Natl Acad Sci U S A 2005; 102:11432-7. [PMID: 16061802 PMCID: PMC1183580 DOI: 10.1073/pnas.0504851102] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Prior analysis has characterized the clonal characteristics of effector CD8(+) T cells specific for the prominent influenza A virus nucleoprotein (NP) and acid polymerase (PA) peptides presented by H2D(b). Using a single-cell approach and determination of CDR3beta profiles, a limited, predominantly "public" repertoire was found for CD8(+)D(b)NP(366)(+)Vbeta8.3+ cells, whereas diverse and "private" T cell antigen receptor (TCR)beta clonotypes were typical of the CD8(+)D(b)PA(224)(+)Vbeta7+ response. This single-cell approach has now been used to relate the contributions of particular clonotypes (or affinities) to high-avidity TCRs, as defined by binding under conditions of limiting tetramer availability. At least by the measure of CDR3beta usage, no difference could be found between total and high-avidity populations in the spectrum of TCR-pMHC affinities throughout the limited, and relatively public, CD8(+)D(b)NP(366)(+)Vbeta8.3+ populations. Conversely, the more even (by clone size), diverse, and private CD8(+)D(b)PA(224)(+)Vbeta7+ response was characterized by the clear partitioning of the largest T cell clones in the high-avidity compartment. These results suggest that the relatively constrained CD8(+)D(b)NP(366)(+)Vbeta8.3+ set utilizes a relatively narrow range of affinities, whereas the broader CD8(+)D(b)PA(224)(+)Vbeta7+ response is induced at a range of TCR-pMHC affinities. Thus, whereas TCR sequence (or affinity) appears to contribute substantially to the avidity profile of diverse virus-specific CD8+ populations, other mechanisms may be prominent where the TCR spectrum is more limited.
Collapse
Affiliation(s)
- Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, Parkville 3010, Melbourne, Australia
| | | | | | | | | |
Collapse
|
147
|
Di Rosa F, Pabst R. The bone marrow: a nest for migratory memory T cells. Trends Immunol 2005; 26:360-6. [PMID: 15978522 DOI: 10.1016/j.it.2005.04.011] [Citation(s) in RCA: 227] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2005] [Revised: 04/12/2005] [Accepted: 04/28/2005] [Indexed: 12/19/2022]
Abstract
It has been known for a long time that T-cell precursors generated in the bone marrow migrate to the thymus, where T-cell development occurs. However, a fact often neglected is that, under physiological conditions, mature CD4 and CD8 lymphocytes undergo extensive migration from the blood to the bone marrow and vice versa. Here, we first review several observations showing that the bone marrow can function as a secondary lymphoid organ for both CD4 and CD8 cells, as well as a preferential homing site for memory T cells. Second, we discuss evidence that, a long time after priming, memory CD8 cells proliferate more extensively in the bone marrow than they do in either secondary lymphoid or extra-lymphoid organs. Finally, we propose that the bone marrow is a central organ in mature T-cell traffic and contributes greatly to long-term cytotoxic memory, which has implications for adoptive immunotherapy and vaccine design.
Collapse
Affiliation(s)
- Francesca Di Rosa
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso, CNR via G. Marconi 10, Naples 80125, Italy.
| | | |
Collapse
|
148
|
Cohen GB, Kaur A, Johnson RP. Isolation of viable antigen-specific CD4 T cells by CD40L surface trapping. J Immunol Methods 2005; 302:103-15. [PMID: 15993419 DOI: 10.1016/j.jim.2005.05.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2004] [Revised: 05/04/2005] [Accepted: 05/04/2005] [Indexed: 11/24/2022]
Abstract
A number of techniques have recently been developed for the identification of antigen-specific cells, yet the ability of these techniques to identify all subclasses of memory T cells has often been overlooked. Here we describe a novel approach for the isolation of live antigen-specific CD4 T cells using CD40L and CD69 surface staining and demonstrate its utility for isolating antigen-specific rhesus macaque CD4 T cells. Critical to the success of the technique was staining for CD40L concurrent with antigen stimulation. Isolation of CD4 T cells based on CD40L/CD69 surface marker upregulation identified both effector and central memory CD4 T cells. In contrast, the majority of central memory CD4 T cells did not secrete TNFalpha or IFNgamma and thus would not be identified by techniques based on their secretion. The methodology described here therefore complements existing approaches for isolating viable antigen-specific CD4 T cells, opens new avenues for investigating human diseases in nonhuman primate animal models and may prove beneficial in instances where the induced response is largely T cell central memory restricted.
Collapse
Affiliation(s)
- George B Cohen
- Department of Biochemistry and Volen Center for Complex Systems, Brandeis University, 415 South Street, Waltham, MA 02454, USA.
| | | | | |
Collapse
|
149
|
Roberts AD, Ely KH, Woodland DL. Differential contributions of central and effector memory T cells to recall responses. ACTA ACUST UNITED AC 2005; 202:123-33. [PMID: 15983064 PMCID: PMC2212898 DOI: 10.1084/jem.20050137] [Citation(s) in RCA: 214] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Although the absolute number of memory CD8+ T cells established in the spleen following antigen encounter remains stable for many years, the relative capacity of these cells to mediate recall responses is not known. Here we used a dual adoptive transfer approach to demonstrate a progressive increase in the quality of memory T cell pools in terms of their ability to proliferate and accumulate at effector sites in response to secondary pathogen challenge. This temporal increase in efficacy occurred in CD62Llo (effector memory) and CD62Lhi (central memory) subpopulations, but was most prominent in the CD62Lhi subpopulation. These data indicate that the contribution of effector memory and central memory T cells to the recall response changes substantially over time.
Collapse
|
150
|
Haring JS, Corbin GA, Harty JT. Dynamic regulation of IFN-gamma signaling in antigen-specific CD8+ T cells responding to infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2005; 174:6791-802. [PMID: 15905520 DOI: 10.4049/jimmunol.174.11.6791] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IFN-gamma plays a critical role in the CD8(+) T cell response to infection, but when and if this cytokine directly signals CD8(+) T cells during an immune response is unknown. We show that naive Ag-specific CD8(+) T cells receive IFN-gamma signals within 12 h after in vivo infection with Listeria monocytogenes and then become unresponsive to IFN-gamma throughout the ensuing Ag-driven expansion phase. Ag-specific CD8(+) T cells regain partial IFN-gamma responsiveness throughout the contraction phase, whereas the memory pool exhibits uniform, but reduced, responsiveness that is also modulated during the secondary response. The responsiveness of Ag-specific CD8(+) T cells to IFN-gamma correlated with modulation in the expression of IFN-gammaR2, but not with IFN-gammaR1 or suppressor of cytokine signaling-1. This dynamic regulation suggests that early IFN-gamma signals participate in regulation of the primary CD8(+) T cell response program, but that evading or minimizing IFN-gamma signals during expansion and the memory phase may contribute to appropriate regulation of the CD8(+) T cell response.
Collapse
Affiliation(s)
- Jodie S Haring
- Department of Microbiology, University of Iowa, Iowa City, 52242, USA
| | | | | |
Collapse
|