51
|
Persistent loss of IL-27 responsiveness in CD8+ memory T cells abrogates IL-10 expression in a recall response. Proc Natl Acad Sci U S A 2012; 109:18535-40. [PMID: 23091017 DOI: 10.1073/pnas.1119133109] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
CD8+ T cells are central to the eradication of intracellular pathogens, but they can also act to limit inflammation and immunopathology. During primary respiratory viral infection CD8+ effector T cells release the immunosuppressive cytokine IL-10, which is essential for host survival. Here we report that CD8+ T-cell-derived IL-10 is absent in a recall response. We show in mice that the lack of IL-10 is due to a persistent loss of IL-27 responsiveness in CD8+ memory T cells, caused by down-regulation of the common cytokine receptor, glycoprotein 130. CD8+ memory T cells secreted less IL-10 when activated in the presence of IL-27 than did naïve controls, and retroviral expression of glycoprotein 130 restored IL-10 and reduced IFN-γ production upon restimulation. We demonstrate that human CD8+ memory cells are also characterized by impaired IL-27 responsiveness. Our data suggest that CD8+ T-cell activation involves a persistent loss of specific cytokine receptors that determines the functional potential of these cells during rechallenge infection.
Collapse
|
52
|
La Gruta N, Kelso A, Brown LE, Chen W, Jackson DC, Turner SJ. Role of CD8(+) T-cell immunity in influenza infection: potential use in future vaccine development. Expert Rev Respir Med 2012; 3:523-37. [PMID: 20477341 DOI: 10.1586/ers.09.44] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Continued circulation of the highly pathogenic avian H5N1 influenza A virus has many people worried that an influenza pandemic is imminent. Compounding this is the realization that H5N1 vaccines based on current influenza vaccine technology (designed to generate protective antibody responses) may be suboptimal at providing protection. As a consequence, there is recent interest in vaccine strategies that elicit cellular immunity, particularly the cytotoxic T lymphocyte response, in an effort to provide protection against a potential pandemic. A major issue is the lack of information about the precise role that these 'hitmen' of the immune system have in protecting against both pandemic and seasonal influenza. We need to know more about how the induction and maintenance of cytotoxic T lymphocytes after influenza infection can impact protection from further infection. The challenge is then to use this information in the design of vaccines that will protect against pandemic influenza and will help optimize CD8(+) killer T-cell responses in other infections.
Collapse
Affiliation(s)
- Nicole La Gruta
- Department of Microbiology and Immunology, The University of Melbourne, Royal Parade, Parkville, Victoria 3010, Australia
| | | | | | | | | | | |
Collapse
|
53
|
Anderson KG, Sung H, Skon CN, Lefrancois L, Deisinger A, Vezys V, Masopust D. Cutting edge: intravascular staining redefines lung CD8 T cell responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2012; 189:2702-6. [PMID: 22896631 PMCID: PMC3436991 DOI: 10.4049/jimmunol.1201682] [Citation(s) in RCA: 249] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Nonlymphoid T cell populations control local infections and contribute to inflammatory diseases, thus driving efforts to understand the regulation of their migration, differentiation, and maintenance. Numerous observations indicate that T cell trafficking and differentiation within the lung are starkly different from what has been described in most nonlymphoid tissues, including intestine and skin. After systemic infection, we found that >95% of memory CD8 T cells isolated from mouse lung via standard methods were actually confined to the pulmonary vasculature, despite perfusion. A respiratory route of challenge increased virus-specific T cell localization within lung tissue, although only transiently. Removing blood-borne cells from analysis by the simple technique of intravascular staining revealed distinct phenotypic signatures and chemokine-dependent trafficking restricted to Ag-experienced T cells. These results precipitate a revised model for pulmonary T cell trafficking and differentiation and a re-evaluation of studies examining the contributions of pulmonary T cells to protection and disease.
Collapse
Affiliation(s)
- Kristin G. Anderson
- Department of Microbiology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Heungsup Sung
- Department of Microbiology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Cara N. Skon
- Department of Microbiology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Leo Lefrancois
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Angela Deisinger
- Department of Microbiology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Vaiva Vezys
- Department of Microbiology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - David Masopust
- Department of Microbiology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
54
|
Wakim LM, Woodward-Davis A, Liu R, Hu Y, Villadangos J, Smyth G, Bevan MJ. The molecular signature of tissue resident memory CD8 T cells isolated from the brain. THE JOURNAL OF IMMUNOLOGY 2012; 189:3462-71. [PMID: 22922816 DOI: 10.4049/jimmunol.1201305] [Citation(s) in RCA: 290] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Tissue resident memory (Trm) CD8 T cells represent a newly described memory T cell population. We have previously characterized a population of Trm cells that persists within the brain after acute virus infection. Although capable of providing marked protection against a subsequent local challenge, brain Trm cells do not undergo recall expansion after dissociation from the tissue. Furthermore, these Trm cells do not depend on the same survival factors as the circulating memory T cell pool as assessed either in vivo or in vitro. To gain greater insight into this population of cells, we compared the gene expression profiles of Trm cells isolated from the brain with those of circulating memory T cells isolated from the spleen after an acute virus infection. Trm cells displayed altered expression of genes involved in chemotaxis, expressed a distinct set of transcription factors, and overexpressed several inhibitory receptors. Cumulatively, these data indicate that Trm cells are a distinct memory T cell population disconnected from the circulating memory T cell pool and display a unique molecular signature that likely results in optimal survival and function within their local environment.
Collapse
Affiliation(s)
- Linda M Wakim
- Department of Immunology, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | | | | |
Collapse
|
55
|
Roy A, Bauer SM, Lawrence BP. Developmental exposure to bisphenol A modulates innate but not adaptive immune responses to influenza A virus infection. PLoS One 2012; 7:e38448. [PMID: 22675563 PMCID: PMC3366985 DOI: 10.1371/journal.pone.0038448] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Accepted: 05/09/2012] [Indexed: 12/25/2022] Open
Abstract
Bisphenol A (BPA) is used in numerous products, such as plastic bottles and food containers, from which it frequently leaches out and is consumed by humans. There is a growing public concern that BPA exposure may pose a significant threat to human health. Moreover, due to the widespread and constant nature of BPA exposure, not only adults but fetuses and neonates are also exposed to BPA. There is mounting evidence that developmental exposures to chemicals from our environment, including BPA, contribute to diseases late in life; yet, studies of how early life exposures specifically alter the immune system are limited. Herein we report an examination of how maternal exposure to a low, environmentally relevant dose of BPA affects the immune response to infection with influenza A virus. We exposed female mice during pregnancy and through lactation to the oral reference dose for BPA listed by the US Environmental Protection Agency, and comprehensively examined immune parameters directly linked to disease outcomes in adult offspring following infection with influenza A virus. We found that developmental exposure to BPA did not compromise disease-specific adaptive immunity against virus infection, or reduce the host's ability to clear the virus from the infected lung. However, maternal exposure to BPA transiently reduced the extent of infection-associated pulmonary inflammation and anti-viral gene expression in lung tissue. From these observations, we conclude that maternal exposure to BPA slightly modulates innate immunity in adult offspring, but does not impair the anti-viral adaptive immune response, which is critical for virus clearance and survival following influenza virus infection.
Collapse
Affiliation(s)
- Anirban Roy
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Stephen M. Bauer
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - B. Paige Lawrence
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| |
Collapse
|
56
|
Long-lived epithelial immunity by tissue-resident memory T (TRM) cells in the absence of persisting local antigen presentation. Proc Natl Acad Sci U S A 2012; 109:7037-42. [PMID: 22509047 DOI: 10.1073/pnas.1202288109] [Citation(s) in RCA: 497] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Although circulating memory T cells provide enhanced protection against pathogen challenge, they often fail to do so if infection is localized to peripheral or extralymphoid compartments. In those cases, it is T cells already resident at the site of virus challenge that offer superior immune protection. These tissue-resident memory T (T(RM)) cells are identified by their expression of the α-chain from the integrin α(E)(CD103)β(7), and can exist in disequilibrium with the blood, remaining in the local environment long after peripheral infections subside. In this study, we demonstrate that long-lived intraepithelial CD103(+)CD8(+) T(RM) cells can be generated in the absence of in situ antigen recognition. Local inflammation in skin and mucosa alone resulted in enhanced recruitment of effector populations and their conversion to the T(RM) phenotype. The CD8(+) T(RM) cells lodged in these barrier tissues provided long-lived protection against local challenge with herpes simplex virus in skin and vagina challenge models, and were clearly superior to the circulating memory T-cell cohort. The results demonstrate that peripheral T(RM) cells can be generated and survive in the absence of local antigen presentation and provide a powerful means of achieving immune protection against peripheral infection.
Collapse
|
57
|
Teijaro JR, Turner D, Pham Q, Wherry EJ, Lefrançois L, Farber DL. Cutting edge: Tissue-retentive lung memory CD4 T cells mediate optimal protection to respiratory virus infection. THE JOURNAL OF IMMUNOLOGY 2011; 187:5510-4. [PMID: 22058417 DOI: 10.4049/jimmunol.1102243] [Citation(s) in RCA: 507] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We identify in this article a new class of lung tissue-resident memory CD4 T cells that exhibit tissue tropism and retention independent of Ag or inflammation. Tissue-resident memory CD4 T cells in the lung did not circulate or emigrate from the lung in parabiosis experiments, were protected from in vivo Ab labeling, and expressed elevated levels of CD69 and CD11a compared with those of circulating memory populations. Importantly, influenza-specific lung-resident memory CD4 T cells served as in situ protectors to respiratory viral challenge, mediating enhanced viral clearance and survival to lethal influenza infection. By contrast, memory CD4 T cells isolated from spleen recirculated among multiple tissues without retention and failed to mediate protection to influenza infection, despite their ability to expand and migrate to the lung. Our results reveal tissue compartmentalization as a major determining factor for immune-mediated protection in a key mucosal site, important for targeting local protective responses in vaccines and immunotherapies.
Collapse
Affiliation(s)
- John R Teijaro
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | | | |
Collapse
|
58
|
Abstract
Bone marrow is thought to be a primary hematopoietic organ. However, accumulated evidences demonstrate that active function and trafficking of immune cells, including regulatory T cells, conventional T cells, B cells, dendritic cells, natural killer T (NKT) cells, neutrophils, myeloid-derived suppressor cells and mesenchymal stem cells, are observed in the bone marrow. Furthermore, bone marrow is a predetermined metastatic location for multiple human tumors. In this review, we discuss the immune network in the bone marrow. We suggest that bone marrow is an immune regulatory organ capable of fine tuning immunity and may be a potential therapeutic target for immunotherapy and immune vaccination.
Collapse
|
59
|
Wu H, Kumar A, Miao H, Holden-Wiltse J, Mosmann TR, Livingstone AM, Belz GT, Perelson AS, Zand MS, Topham DJ. Modeling of influenza-specific CD8+ T cells during the primary response indicates that the spleen is a major source of effectors. THE JOURNAL OF IMMUNOLOGY 2011; 187:4474-82. [PMID: 21948988 DOI: 10.4049/jimmunol.1101443] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The biological parameters that determine the distribution of virus-specific CD8(+) T cells during influenza infection are not all directly measurable by experimental techniques but can be inferred through mathematical modeling. Mechanistic and semimechanistic ordinary differential equations were developed to describe the expansion, trafficking, and disappearance of activated virus-specific CD8(+) T cells in lymph nodes, spleens, and lungs of mice during primary influenza A infection. An intensive sampling of virus-specific CD8(+) T cells from these three compartments was used to inform the models. Rigorous statistical fitting of the models to the experimental data allowed estimation of important biological parameters. Although the draining lymph node is the first tissue in which Ag-specific CD8(+) T cells are detected, it was found that the spleen contributes the greatest number of effector CD8(+) T cells to the lung, with rates of expansion and migration that exceeded those of the draining lymph node. In addition, models that were based on the number and kinetics of professional APCs fit the data better than those based on viral load, suggesting that the immune response is limited by Ag presentation rather than the amount of virus. Modeling also suggests that loss of effector T cells from the lung is significant and time dependent, increasing toward the end of the acute response. Together, these efforts provide a better understanding of the primary CD8(+) T cell response to influenza infection, changing the view that the spleen plays a minor role in the primary immune response.
Collapse
Affiliation(s)
- Hulin Wu
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Neutrophils sustain effective CD8(+) T-cell responses in the respiratory tract following influenza infection. Immunol Cell Biol 2011; 90:197-205. [PMID: 21483446 DOI: 10.1038/icb.2011.26] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Neutrophils have an important role in early host protection during influenza A virus infection. Their ability to modulate the virus-specific adaptive immune response is less clear. Here, we have used a mouse model to examine the impact of neutrophils on CD8(+) T-cell responses during influenza virus infection. CD8(+) T-cell priming, expansion, migration, cytokine secretion and cytotoxic capacity were investigated in the virus-infected airways and secondary lymphoid organs. To do this, we utilised a Ly6G-specific monoclonal antibody (mAb; 1A8) that specifically depletes neutrophils in vivo. Neutrophil depletion early after infection with influenza virus strain HKx31 (H3N2) did not alter influenza virus-derived antigen presentation or naïve CD8(+) T-cell expansion in the secondary lymphoid organs. Trafficking of virus-specific CD8(+) T cells into the infected pulmonary airways was also unaltered. Instead, early neutropenia reduced both the overall magnitude of influenza virus-specific CD8(+) T cells, together with impaired cytokine production and cytotoxic effector function. Therefore, neutrophils are important participants in anti-viral mechanisms that sustain effective CD8(+) T-cell responses in the respiratory tract of influenza virus-infected mice.
Collapse
|
61
|
Abstract
CD4+ T cells - often referred to as T-helper cells - play a central role in immune defense and pathogenesis. Virus infections and vaccines stimulate and expand populations of antigen-specific CD4+ T cells in mice and in man. These virus-specific CD4+ T cells are extremely important in antiviral protection: deficiencies in CD4+ T cells are associated with virus reactivation, generalized susceptibility to opportunistic infections, and poor vaccine efficacy. As described below, CD4+ T cells influence effector and memory CD8+ T cell responses, humoral immunity, and the antimicrobial activity of macrophages and are involved in recruiting cells to sites of infection. This review summarizes a few key points about the dynamics of the CD4+ T cell response to virus infection, the positive role of pro-inflammatory cytokines in the differentiation of virus-specific CD4+ T cells, and new areas of investigation to improve vaccines against virus infection.
Collapse
Affiliation(s)
- Jason K Whitmire
- Carolina Vaccine Institute, The University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
62
|
Environmental and antigen receptor-derived signals support sustained surveillance of the lungs by pathogen-specific cytotoxic T lymphocytes. J Virol 2011; 85:4085-94. [PMID: 21345961 DOI: 10.1128/jvi.02493-10] [Citation(s) in RCA: 149] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Viral infections often gain access to the body of their host by exploiting areas of natural vulnerability, such as the semipermeable surfaces of mucosal tissues which are adapted for adsorption of nutrients and other diffusible molecules. Once the microbes have crossed the epithelial barrier, they can disperse to other tissues where eradication may not be possible. The best opportunity for successful immune intervention is immediately after infection while the pathogen is confined to a localized area of the body. Cytotoxic T lymphocytes (CTL) which reside at the site where the infection begins can make an important contribution to immunity by reducing early dissemination of the infection. Because the lungs provide easy access points for many pathogens to enter the body, they require protection from many complementary mechanisms, including pathogen-specific cytotoxic T cells. In this study we show that an enduring response to pathogen-derived peptide antigens facilitates sustained surveillance of the lungs by pathogen-specific CTL during the recovery from influenza virus infection. Our studies show that these processed peptide antigens reinforce expression of two homing receptors (CD69 and CD103) which help recently activated virus-specific CTL colonize the lungs during a mild inflammatory response. We suggest that this requirement for prolonged antigen presentation to reinforce local CTL responses in the lungs explains why protective cellular immunity quickly declines following influenza virus infection and other viral infections that enter the body via mucosal tissues.
Collapse
|
63
|
Croom HA, Denton AE, Valkenburg SA, Swan NG, Olson MR, Turner SJ, Doherty PC, Kedzierska K. Memory precursor phenotype of CD8+ T cells reflects early antigenic experience rather than memory numbers in a model of localized acute influenza infection. Eur J Immunol 2011; 41:682-93. [PMID: 21264852 DOI: 10.1002/eji.201040625] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Revised: 11/09/2010] [Accepted: 12/07/2010] [Indexed: 01/03/2023]
Abstract
The mechanistic basis of memory T-cell development is poorly defined. Phenotypic markers that define precursors at effector stages have been characterized for acute systemic infections with high antigen load. We asked whether such markers can identify memory precursors from early effectors (d6) to late memory (>d500) for two immunodominant CD8(+) responses during the course of a localized low-load influenza infection in mice. CD8(+) T cells stained with the D(b) NP(366) and D(b) PA(224) tetramers were characterized as IL-7Rα(hi) , IL-7Rα(hi) CD62L(hi) or IL-7Rα(hi) KLRG1(lo) . While the D(b) NP(366) - and D(b) PA(224) -specific responses were comparable in size, decay kinetics and memory precursor frequency, their expansion characteristics differed. This correlated with a divergence in the IL-7Rα(hi) , IL-7Rα(hi) CD62L(hi) and IL-7Rα(hi) KLRG1(lo) phenotypes on effector, but not naïve, CD8(+) populations. That effect was abrogated by priming with viruses engineered to present equivalent levels of NP(366) and PA(224) peptides, indicating that memory phenotypes reflect early antigenic experience rather than memory potential. Thus, the IL-7Rα(hi) KLRG1(lo) phenotype had a poor predictive value in identifying memory precursors in the spleen and at the site of infection. Greater consistency in influenza-specific IL-7Rα(hi) KLRG1(lo) CD8(+) T-cell numbers was found in draining lymph nodes, suggesting that this may be the preferential site for memory establishment and maintenance following localized virus infections.
Collapse
Affiliation(s)
- Hayley A Croom
- Department of Microbiology and Immunology, University of Melbourne, Parkville, Melbourne, Australia
| | | | | | | | | | | | | | | |
Collapse
|
64
|
Selin LK, Wlodarczyk MF, Kraft AR, Nie S, Kenney LL, Puzone R, Celada F. Heterologous immunity: immunopathology, autoimmunity and protection during viral infections. Autoimmunity 2011; 44:328-47. [PMID: 21250837 DOI: 10.3109/08916934.2011.523277] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Heterologous immunity is a common phenomenon present in all infections. Most of the time it is beneficial, mediating protective immunity, but in some individuals that have the wrong crossreactive response it leads to a cascade of events that result in severe immunopathology. Infections have been associated with autoimmune diseases such as diabetes, multiple sclerosis and lupus erythematosis, but also with unusual autoimmune like pathologies where the immune system appears dysregulated, such as, sarcoidosis, colitis, panniculitis, bronchiolitis obliterans, infectious mononucleosis and even chronic fatigue syndrome. Here we review the evidence that to better understand these autoreactive pathologies it requires an evaluation of how T cells are regulated and evolve during sequential infections with different pathogens under the influence of heterologous immunity.
Collapse
Affiliation(s)
- Liisa K Selin
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | | | | | | | | | | | | |
Collapse
|
65
|
Vaccine protection against lethal homologous and heterologous challenge using recombinant AAV vectors expressing codon-optimized genes from pandemic swine origin influenza virus (SOIV). Vaccine 2010; 29:1690-9. [PMID: 21195079 DOI: 10.1016/j.vaccine.2010.12.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2010] [Revised: 08/16/2010] [Accepted: 12/14/2010] [Indexed: 12/12/2022]
Abstract
The recent H1N1 influenza pandemic and the inevitable delay between identification of the virus and production of the specific vaccine have highlighted the urgent need for new generation influenza vaccines that can preemptively induce broad immunity to different strains of the virus. In this study we have produced AAV-based vectors expressing the A/Mexico/4603/2009 (H1N1) hemagglutinin (HA), nucleocapsid (NP) and the matrix protein M1 and have evaluated their ability to induce specific immune response and protect mice against homologous and heterologous challenge. Each of the vaccine vectors elicited potent cellular and humoral immune responses in mice. Although immunization with AAV-M1 did not improve survival after challenge with the homologous strain, immunization with the AAV-H1 and AAV-NP vectors resulted in survival of all mice, as did inoculation with a combination of all three vectors. Furthermore, trivalent vaccination also conferred partial protection against challenge with the highly heterologous and virulent A/PR/8/34 strain of H1N1 influenza.
Collapse
|
66
|
McComb S, Mulligan R, Sad S. Caspase-3 is transiently activated without cell death during early antigen driven expansion of CD8(+) T cells in vivo. PLoS One 2010; 5:e15328. [PMID: 21203525 PMCID: PMC3008739 DOI: 10.1371/journal.pone.0015328] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Accepted: 11/08/2010] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND CD8(+) T cell responses develop rapidly during infection and are swiftly reduced during contraction, wherein >90% of primed CD8(+) T cells are eliminated. The role of apoptotic mechanisms in controlling this rapid proliferation and contraction of CD8(+) T cells remains unclear. Surprisingly, evidence has shown non-apoptotic activation of caspase-3 to occur during in vitro T-cell proliferation, but the relevance of these mechanisms to in vivo CD8(+) T cell responses has yet to be examined. METHODS AND FINDINGS We have evaluated the activity of caspase-3, a key downstream inducer of apoptosis, throughout the entirety of a CD8(+) T cell response. We utilized two infection models that differ in the intensity, onset and duration of antigen-presentation and inflammation. Expression of cleaved caspase-3 in antigen specific CD8(+) T cells was coupled to the timing and strength of antigen presentation in lymphoid organs. We also observed coordinated activation of additional canonical apoptotic markers, including phosphatidylserine exposure. Limiting dilution analysis directly showed that in the presence of IL7, very little cell death occurred in both caspase-3(hi) and caspase-3(low) CD8(+) T cells. The expression of active caspase-3 peaked before effector phenotype (CD62L(low)) CD8(+) T cells emerged, and was undetectable in effector-phenotype cells. In addition, OVA-specific CD8(+) cells remained active caspase-3(low) throughout the contraction phase. CONCLUSIONS Our results specifically implicate antigen and not inflammation in driving activation of apoptotic mechanisms without cell death in proliferating CD8(+) T cells. Furthermore, the contraction of CD8(+) T cell response following expansion is likely not mediated by the key downstream apoptosis inducer, caspase-3.
Collapse
Affiliation(s)
- Scott McComb
- NRC-Institute for Biological Sciences, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada
| | - Rebecca Mulligan
- NRC-Institute for Biological Sciences, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada
| | - Subash Sad
- NRC-Institute for Biological Sciences, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada
| |
Collapse
|
67
|
Wakim LM, Woodward-Davis A, Bevan MJ. Memory T cells persisting within the brain after local infection show functional adaptations to their tissue of residence. Proc Natl Acad Sci U S A 2010; 107:17872-9. [PMID: 20923878 PMCID: PMC2964240 DOI: 10.1073/pnas.1010201107] [Citation(s) in RCA: 437] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The brain is not routinely surveyed by lymphocytes and is defined as an immuno-privileged site. However, viral infection of the brain results in the infiltration and long-term persistence of pathogen-specific CD8(+) T cells. These cells survive without replenishment from the circulation and are referred to as resident memory T cells (Trm). Brain Trm selectively express the integrin CD103, the expression of which is dependent on antigen recognition within the tissue. After clearance of virus, CD8(+) T cells persist in tight clusters, presumably at prior infection hot spots. Antigen persistence is not a prerequisite for T-cell retention, as suggested by the failure to detect viral genomes in the T-cell clusters. Furthermore, we show that an intracranial dendritic cell immunization regimen, which allows the transient introduction of antigen, also results in the generation of memory T cells that persist long term in the brain. Brain Trm die rapidly on isolation from the tissue and fail to undergo recall expansion after adoptive transfer into the bloodstream of antigen-challenged recipients. These ex vivo defects imply a dependency on the local milieu for function and survival. Cumulatively, this work shows that Trm are a specialized population of memory T cells that can be deposited in tissues previously thought to be beyond routine immune surveillance.
Collapse
Affiliation(s)
- Linda M. Wakim
- Department of Immunology, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195
| | - Amanda Woodward-Davis
- Department of Immunology, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195
| | - Michael J. Bevan
- Department of Immunology, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195
| |
Collapse
|
68
|
Kim TS, Hufford MM, Sun J, Fu YX, Braciale TJ. Antigen persistence and the control of local T cell memory by migrant respiratory dendritic cells after acute virus infection. J Exp Med 2010; 207:1161-72. [PMID: 20513748 PMCID: PMC2882836 DOI: 10.1084/jem.20092017] [Citation(s) in RCA: 155] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2009] [Accepted: 05/12/2010] [Indexed: 11/30/2022] Open
Abstract
Acute viral infections induce robust adaptive immune responses resulting in virus clearance. Recent evidence suggests that there may be depots of viral antigen that persist in draining lymph nodes (DLNs) after virus clearance and could, therefore, affect the adaptive immune response and memory T cell formation. The nature of these residual antigen depots, the mechanism of antigen persistence, and the impact of the persistent antigen on memory T cells remain ill defined. Using a mouse model of influenza virus infection of the respiratory tract, we identified respiratory dendritic cells (RDCs) as essential for both sampling and presenting residual viral antigen. RDCs in the previously infected lung capture residual viral antigen deposited in an irradiation-resistant cell type. RDCs then transport the viral antigen to the LNs draining the site of infection, where they present the antigen to T cells. Lastly, we document preferential localization of memory T cells to the DLNs after virus clearance as a consequence of presentation of residual viral antigen by the migrant RDC.
Collapse
Affiliation(s)
- Taeg S. Kim
- Beirne B. Carter Center for Immunology Research, Department of Microbiology, and Department of Pathology, University of Virginia, Charlottesville, VA 22908
| | - Matthew M. Hufford
- Beirne B. Carter Center for Immunology Research, Department of Microbiology, and Department of Pathology, University of Virginia, Charlottesville, VA 22908
| | - Jie Sun
- Beirne B. Carter Center for Immunology Research, Department of Microbiology, and Department of Pathology, University of Virginia, Charlottesville, VA 22908
| | - Yang-Xin Fu
- Department of Pathology, University of Chicago, Chicago, IL 60637
| | - Thomas J. Braciale
- Beirne B. Carter Center for Immunology Research, Department of Microbiology, and Department of Pathology, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
69
|
Wakim LM, Bevan MJ. From the thymus to longevity in the periphery. Curr Opin Immunol 2010; 22:274-8. [PMID: 20378321 PMCID: PMC2904522 DOI: 10.1016/j.coi.2010.03.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2010] [Accepted: 03/07/2010] [Indexed: 11/30/2022]
Abstract
An important attribute of the adaptive immune system is the ability to remember a prior encounter with a pathogen; an ability termed immunological memory. Bigger, better, and stronger responses are mounted upon a secondary encounter with the pathogen potentially resulting in clearance of the infection before the development of disease. We will review recent advances in the field of memory CD8(+) T cell differentiation focusing on both intrinsic and extrinsic factors that govern the development of T cell memory.
Collapse
Affiliation(s)
- Linda M Wakim
- Department of Immunology, Howard Hughes Medical Institute, University of Washington, Box 357370, Seattle, Washington 98195, USA
| | - Michael J Bevan
- Department of Immunology, Howard Hughes Medical Institute, University of Washington, Box 357370, Seattle, Washington 98195, USA
| |
Collapse
|
70
|
Masopust D, Choo D, Vezys V, Wherry EJ, Duraiswamy J, Akondy R, Wang J, Casey KA, Barber DL, Kawamura KS, Fraser KA, Webby RJ, Brinkmann V, Butcher EC, Newell KA, Ahmed R. Dynamic T cell migration program provides resident memory within intestinal epithelium. J Exp Med 2010; 207:553-64. [PMID: 20156972 PMCID: PMC2839151 DOI: 10.1084/jem.20090858] [Citation(s) in RCA: 497] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Accepted: 01/13/2010] [Indexed: 01/01/2023] Open
Abstract
Migration to intestinal mucosa putatively depends on local activation because gastrointestinal lymphoid tissue induces expression of intestinal homing molecules, whereas skin-draining lymph nodes do not. This paradigm is difficult to reconcile with reports of intestinal T cell responses after alternative routes of immunization. We reconcile this discrepancy by demonstrating that activation within spleen results in intermediate induction of homing potential to the intestinal mucosa. We further demonstrate that memory T cells within small intestine epithelium do not routinely recirculate with memory T cells in other tissues, and we provide evidence that homing is similarly dynamic in humans after subcutaneous live yellow fever vaccine immunization. These data explain why systemic immunization routes induce local cell-mediated immunity within the intestine and indicate that this tissue must be seeded with memory T cell precursors shortly after activation.
Collapse
Affiliation(s)
- David Masopust
- Department of Microbiology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455
- Emory Vaccine Center and Department of Microbiology and Immunology and Department of Pathology, Emory University School of Medicine, Atlanta GA 30322
| | - Daniel Choo
- Emory Vaccine Center and Department of Microbiology and Immunology and Department of Pathology, Emory University School of Medicine, Atlanta GA 30322
| | - Vaiva Vezys
- Department of Microbiology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455
- Emory Vaccine Center and Department of Microbiology and Immunology and Department of Pathology, Emory University School of Medicine, Atlanta GA 30322
| | - E. John Wherry
- Emory Vaccine Center and Department of Microbiology and Immunology and Department of Pathology, Emory University School of Medicine, Atlanta GA 30322
| | - Jaikumar Duraiswamy
- Emory Vaccine Center and Department of Microbiology and Immunology and Department of Pathology, Emory University School of Medicine, Atlanta GA 30322
| | - Rama Akondy
- Emory Vaccine Center and Department of Microbiology and Immunology and Department of Pathology, Emory University School of Medicine, Atlanta GA 30322
| | - Jun Wang
- Emory Vaccine Center and Department of Microbiology and Immunology and Department of Pathology, Emory University School of Medicine, Atlanta GA 30322
| | - Kerry A. Casey
- Department of Microbiology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Daniel L. Barber
- Emory Vaccine Center and Department of Microbiology and Immunology and Department of Pathology, Emory University School of Medicine, Atlanta GA 30322
| | - Kim S. Kawamura
- Emory Vaccine Center and Department of Microbiology and Immunology and Department of Pathology, Emory University School of Medicine, Atlanta GA 30322
| | - Kathryn A. Fraser
- Department of Microbiology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Richard J. Webby
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Volker Brinkmann
- Autoimmunity, Transplantation, and Inflammation, Novartis Institutes for Biomedical Research, CH-4002 Basel, Switzerland
| | - Eugene C. Butcher
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305
| | - Kenneth A. Newell
- Emory Vaccine Center and Department of Microbiology and Immunology and Department of Pathology, Emory University School of Medicine, Atlanta GA 30322
| | - Rafi Ahmed
- Emory Vaccine Center and Department of Microbiology and Immunology and Department of Pathology, Emory University School of Medicine, Atlanta GA 30322
| |
Collapse
|
71
|
Karlsson EA, Sheridan PA, Beck MA. Diet-induced obesity impairs the T cell memory response to influenza virus infection. THE JOURNAL OF IMMUNOLOGY 2010; 184:3127-33. [PMID: 20173021 DOI: 10.4049/jimmunol.0903220] [Citation(s) in RCA: 177] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The Centers for Disease Control and Prevention has suggested that obesity may be an independent risk factor for increased severity of illness from the H1N1 pandemic strain. Memory T cells generated during primary influenza infection target internal proteins common among influenza viruses, making them effective against encounters with heterologous strains. In male, diet-induced obese C57BL/6 mice, a secondary H1N1 influenza challenge following a primary H3N2 infection led to a 25% mortality rate (with no loss of lean controls), 25% increase in lung pathology, failure to regain weight, and 10- to 100-fold higher lung viral titers. Furthermore, mRNA expression for IFN-gamma was >60% less in lungs of obese mice, along with one third the number of influenza-specific CD8(+) T cells producing IFN-gamma postsecondary infection versus lean controls. Memory CD8(+) T cells from obese mice had a >50% reduction in IFN-gamma production when stimulated with influenza-pulsed dendritic cells from lean mice. Thus, the function of influenza-specific memory T cells is significantly reduced and ineffective in lungs of obese mice. The reality of a worldwide obesity epidemic combined with yearly influenza outbreaks and the current pandemic makes it imperative to understand how influenza virus infection behaves differently in an obese host. Moreover, impairment of memory responses has significant implications for vaccine efficacy in an obese population.
Collapse
Affiliation(s)
- Erik A Karlsson
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | |
Collapse
|
72
|
|
73
|
CD8(+) T lymphocyte mobilization to virus-infected tissue requires CD4(+) T-cell help. Nature 2009; 462:510-3. [PMID: 19898495 PMCID: PMC2789415 DOI: 10.1038/nature08511] [Citation(s) in RCA: 446] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Accepted: 09/16/2009] [Indexed: 01/17/2023]
Abstract
CD4+ T helper cells are well known for their role in providing critical signals during priming of cytotoxic CD8+ T lymphocyte (CTL) responses in vivo. T help is required for the generation of primary CTL responses as well as in promoting protective CD8+ memory T cell development1. However, the role of CD4 help in the control of CTL responses at the effector stage is unknown. Here, we show that fully helped effector CTLs are not themselves self-sufficient for entry into the infected tissue, but rely on the CD4+ T cells to provide the necessary cue. CD4+ T helper cells control the migration of CTL indirectly through the secretion of IFN-γ and induction of local chemokine secretion in the infected tissue. Our results reveal a previously unappreciated role of CD4 help in mobilizing effector CTL to the peripheral sites of infection where they help to eliminate infected cells.
Collapse
|
74
|
Gebhardt T, Carbone FR. Immunology: A helpers' guide to infection. Nature 2009. [DOI: 10.1038/nature08606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
75
|
Vaccination focusing immunity on conserved antigens protects mice and ferrets against virulent H1N1 and H5N1 influenza A viruses. Vaccine 2009; 27:6512-21. [DOI: 10.1016/j.vaccine.2009.08.053] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Revised: 08/07/2009] [Accepted: 08/17/2009] [Indexed: 12/14/2022]
|
76
|
Macallan DC, Asquith B, Zhang Y, de Lara C, Ghattas H, Defoiche J, Beverley PCL. Measurement of proliferation and disappearance of rapid turnover cell populations in human studies using deuterium-labeled glucose. Nat Protoc 2009; 4:1313-27. [DOI: 10.1038/nprot.2009.117] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
77
|
Maue AC, Yager EJ, Swain SL, Woodland DL, Blackman MA, Haynes L. T-cell immunosenescence: lessons learned from mouse models of aging. Trends Immunol 2009; 30:301-5. [PMID: 19541537 PMCID: PMC3755270 DOI: 10.1016/j.it.2009.04.007] [Citation(s) in RCA: 131] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Revised: 04/16/2009] [Accepted: 04/17/2009] [Indexed: 01/08/2023]
Abstract
It is well established that increasing age is associated with a decreased capacity of the immune system to mediate effective immune responses to vaccination and invading pathogens. Because of the inherent limitations of conducting experiments in humans, much of what we have learned is owed to the utility of experimental mouse models of aging. Recent studies performed in the mouse have demonstrated mechanisms responsible for age-related declines in the function of CD4(+) and CD8(+) cells. This review describes key findings regarding age-related defects in T-cell function and discusses the impact these defects have on vaccine efficacy and immunity.
Collapse
|
78
|
Kreijtz JHCM, Bodewes R, van den Brand JMA, de Mutsert G, Baas C, van Amerongen G, Fouchier RAM, Osterhaus ADME, Rimmelzwaan GF. Infection of mice with a human influenza A/H3N2 virus induces protective immunity against lethal infection with influenza A/H5N1 virus. Vaccine 2009; 27:4983-9. [PMID: 19538996 DOI: 10.1016/j.vaccine.2009.05.079] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Revised: 05/07/2009] [Accepted: 05/28/2009] [Indexed: 10/20/2022]
Abstract
The transmission of highly pathogenic avian influenza (HPAI) A viruses of the H5N1 subtype from poultry to man and the high case fatality rate fuels the fear for a pandemic outbreak caused by these viruses. However, prior infections with seasonal influenza A/H1N1 and A/H3N2 viruses induce heterosubtypic immunity that could afford a certain degree of protection against infection with the HPAI A/H5N1 viruses, which are distantly related to the human influenza A viruses. To assess the protective efficacy of such heterosubtypic immunity mice were infected with human influenza virus A/Hong Kong/2/68 (H3N2) 4 weeks prior to a lethal infection with HPAI virus A/Indonesia/5/05 (H5N1). Prior infection with influenza virus A/Hong Kong/2/68 reduced clinical signs, body weight loss, mortality and virus replication in the lungs as compared to naive mice infected with HPAI virus A/Indonesia/5/05. Priming by infection with respiratory syncytial virus, a non-related virus did not have a beneficial effect on the outcome of A/H5N1 infections, indicating that adaptive immune responses were responsible for the protective effect. In mice primed by infection with influenza A/H3N2 virus cytotoxic T lymphocytes (CTL) specific for NP(366-374) epitope ASNENMDAM and PA(224-232) SCLENFRAYV were observed. A small proportion of these CTL was cross-reactive with the peptide variant derived from the influenza A/H5N1 virus (ASNENMEVM and SSLENFRAYV respectively) and upon challenge infection with the influenza A/H5N1 virus cross-reactive CTL were selectively expanded. These CTL, in addition to those directed to conserved epitopes, shared by the influenza A/H3N2 and A/H5N1 viruses, most likely contributed to accelerated clearance of the influenza A/H5N1 virus infection. Although also other arms of the adaptive immune response may contribute to heterosubtypic immunity, the induction of virus-specific CTL may be an attractive target for development of broad protective vaccines. Furthermore the existence of pre-existing heterosubtypic immunity may dampen the impact a future influenza pandemic may have.
Collapse
Affiliation(s)
- J H C M Kreijtz
- Department of Virology, Erasmus Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Abstract
The respiratory tract is characterized by an extensive surface area that is in direct contact with the environment, posing a significant problem for effective immune surveillance. Yet most respiratory pathogens are quickly recognized and controlled by a coordinated response involving the innate and adaptive arms of the immune system. The investigation of pulmonary immunity to respiratory viruses during a primary infection has demonstrated that multiple innate and adaptive immune mechanisms are necessary for efficient antiviral responses, and the inhibition of any single mechanism can have disastrous consequences for the host. Furthermore, the investigation of recall responses in the lung has shown that protection from a secondary challenge infection is a complex and elegant process that occurs in distinct stages. In this review, we discuss recent advances that describe the roles of individual components during primary and secondary responses to respiratory virus infections and how these discoveries have added to our understanding of antiviral immunity in the lung.
Collapse
|
80
|
WIESEL MELANIE, WALTON SENTA, RICHTER KIRSTEN, OXENIUS ANNETTE. Virus-specific CD8 T cells: activation, differentiation and memory formation. APMIS 2009; 117:356-81. [DOI: 10.1111/j.1600-0463.2009.02459.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
81
|
Mintern JD, Bedoui S, Davey GM, Moffat JM, Doherty PC, Turner SJ. Transience of MHC Class I-restricted antigen presentation after influenza A virus infection. Proc Natl Acad Sci U S A 2009; 106:6724-9. [PMID: 19346476 PMCID: PMC2672519 DOI: 10.1073/pnas.0901128106] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2009] [Indexed: 11/18/2022] Open
Abstract
Antigen expressed as MHC Class I glycoprotein (pMHCI) complexes on dendritic cells is the primary driver of CD8(+) T cell clonal expansion and differentiation. As we seek to define the molecular differences between acutely stimulated cytotoxic T lymphocyte (CTL) effectors and long-lived memory T cells, it is essential that we understand the duration of in vivo pMHCI persistence. Although infectious influenza A virus is readily cleared by mammalian hosts, that does not necessarily mean that all influenza antigen is totally eliminated. An exhaustive series of carefully controlled adoptive transfer experiments using 3 different carboxy fluorescein diacetate succinimidyl ester-labeled T cell receptor-transgenic CTL populations and a spectrum of genetically engineered and wild-type influenza A viruses provided no evidence for pMHCI persistence over the 30-60-d interval after virus challenge. Molecular profiles identified in antigen-specific T cells at this time may thus be considered to reflect established immunologic memory and not recent CTL activation from a persistent pMHCI pool.
Collapse
Affiliation(s)
- Justine D. Mintern
- Department of Microbiology and Immunology, University of Melbourne, Parkville 3010, Australia
| | - Sammy Bedoui
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3050, Australia
| | - Gayle M. Davey
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3050, Australia
| | - Jessica M. Moffat
- Department of Microbiology and Immunology, University of Melbourne, Parkville 3010, Australia
| | - Peter C. Doherty
- Department of Microbiology and Immunology, University of Melbourne, Parkville 3010, Australia
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105; and
| | - Stephen J. Turner
- Department of Microbiology and Immunology, University of Melbourne, Parkville 3010, Australia
| |
Collapse
|
82
|
Woodland DL, Kohlmeier JE. Migration, maintenance and recall of memory T cells in peripheral tissues. Nat Rev Immunol 2009; 9:153-61. [PMID: 19240755 DOI: 10.1038/nri2496] [Citation(s) in RCA: 315] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
After the resolution of an immune response, antigen-specific memory T cells persist at many sites in the body. The antigen-specific memory T-cell pool includes memory T cells that preferentially reside in peripheral tissues, such as the skin, gut and lungs, where they provide a first line of defence against secondary pathogen infection. Determining how peripheral memory T cells are regulated is essential for our understanding of host-pathogen interactions and for vaccine development. In this Review, we discuss recent insights into the generation, control and recall of peripheral T-cell memory responses.
Collapse
|
83
|
Melenhorst JJ, Scheinberg P, Chattopadhyay PK, Gostick E, Ladell K, Roederer M, Hensel NF, Douek DC, Barrett AJ, Price DA. High avidity myeloid leukemia-associated antigen-specific CD8+ T cells preferentially reside in the bone marrow. Blood 2009; 113:2238-44. [PMID: 18997173 PMCID: PMC2652369 DOI: 10.1182/blood-2008-04-151969] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2008] [Accepted: 10/18/2008] [Indexed: 01/03/2023] Open
Abstract
The activity of allogeneic CD8(+) T cells specific for leukemia-associated antigens (LAAs) is thought to mediate, at least in part, the curative effects of hematopoietic stem cell transplantation (HSCT) in myeloid malignancies. However, the identity and nature of clinically relevant LAA-specific CD8(+) T-cell populations have proven difficult to define. Here, we used a combination of coreceptor-mutated peptide-major histocompatibility complex class I (pMHCI) tetramers and polychromatic flow cytometry to examine the avidity profiles, phenotypic characteristics, and anatomical distribution of HLA A*0201-restricted CD8(+) T-cell populations specific for LAAs that are over-expressed in myeloid leukemias. Remarkably, LAA-specific CD8(+) T-cell populations, regardless of fine specificity, were confined almost exclusively to the bone marrow; in contrast, CD8(+) T-cell populations specific for the HLA A*0201-restricted cytomegalovirus (CMV) pp65(495-503) epitope were phenotypically distinct and evenly distributed between bone marrow and peripheral blood. Furthermore, bone marrow-resident LAA-specific CD8(+) T cells frequently engaged cognate antigen with high avidity; notably, this was the case in all tested bone marrow samples derived from patients who achieved clinical remission after HSCT. These data suggest that concomitant examination of bone marrow specimens in patients with myeloid leukemias might yield more definitive information in the search for immunologic prognosticators of clinical outcome.
Collapse
Affiliation(s)
- J Joseph Melenhorst
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Mansoor N, Abel B, Scriba TJ, Hughes J, de Kock M, Tameris M, Mlenjeni S, Denation L, Little F, Gelderbloem S, Hawkridge A, Boom WH, Kaplan G, Hussey GD, Hanekom WA. Significantly skewed memory CD8+ T cell subsets in HIV-1 infected infants during the first year of life. Clin Immunol 2009; 130:280-9. [PMID: 18996749 PMCID: PMC2722743 DOI: 10.1016/j.clim.2008.09.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2008] [Revised: 09/01/2008] [Accepted: 09/10/2008] [Indexed: 01/20/2023]
Abstract
HIV-1 infection causes a severe T cell compromise; however, little is known about changes in naive, memory, effector and senescent T cell subsets during the first year of life. T cell subsets were studied over the first year of life in blood from 3 infant cohorts: untreated HIV-infected, HIV-exposed but uninfected, and HIV-unexposed. In HIV-infected infants, the frequency of CCR7(+)CD45RA(+) naive CD8(+) T cells was significantly decreased, while the frequency of CCR7(-)CD45RA(-) effector memory CD8(+) T cells was increased, compared with the control cohorts. A larger population of CD8(+) T cells in HIV-infected infants displayed a phenotype consistent with senescence. Differences in CD4(+) T cell subset frequencies were less pronounced, and no significant differences were observed between exposed and unexposed HIV-uninfected infants. We concluded that the proportion of naive, memory, effector and senescent CD8(+) T cells during the first year of life is significantly altered by HIV-1 infection.
Collapse
Affiliation(s)
- Nazma Mansoor
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Anzio Road, Observatory, 7925, South Africa
| | - Brian Abel
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Anzio Road, Observatory, 7925, South Africa
| | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Anzio Road, Observatory, 7925, South Africa
| | - Jane Hughes
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Anzio Road, Observatory, 7925, South Africa
| | - Marwou de Kock
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Anzio Road, Observatory, 7925, South Africa
| | - Michele Tameris
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Anzio Road, Observatory, 7925, South Africa
| | - Sylvia Mlenjeni
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Anzio Road, Observatory, 7925, South Africa
| | - Lea Denation
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Anzio Road, Observatory, 7925, South Africa
| | - Francesca Little
- Department of Statistical Sciences, University of Cape Town, Rondebosch, South Africa
| | - Sebastian Gelderbloem
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Anzio Road, Observatory, 7925, South Africa
| | - Anthony Hawkridge
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Anzio Road, Observatory, 7925, South Africa
| | - W. Henry Boom
- Tuberculosis Research Unit, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, Ohio, USA
| | - Gilla Kaplan
- Public Health Research Institute, University of Medicine and Dentistry of New Jersey, Newark, New Jersey, USA
| | - Gregory D. Hussey
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Anzio Road, Observatory, 7925, South Africa
| | - Willem A. Hanekom
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine and School of Child and Adolescent Health, University of Cape Town, Anzio Road, Observatory, 7925, South Africa
| |
Collapse
|
85
|
Hutchison S, Choo-Kang BSW, Gibson VB, Bundick RV, Leishman AJ, Brewer JM, McInnes IB, Garside P. An investigation of the impact of the location and timing of antigen-specific T cell division on airways inflammation. Clin Exp Immunol 2009; 155:107-16. [PMID: 19076834 DOI: 10.1111/j.1365-2249.2008.03800.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
It is widely accepted that allergic asthma is orchestrated by T helper type 2 lymphocytes specific for inhaled allergen. However, it remains unclear where and when T cell activation and division occurs after allergen challenge, and whether these factors have a significant impact on airways inflammation. We therefore employed a CD4-T cell receptor transgenic adoptive transfer model in conjunction with laser scanning cytometry to characterize the location and timing of T cell division in asthma in vivo. Thus, for the first time we have directly assessed the division of antigen-specific T cells in situ. We found that accumulation of divided antigen-specific T cells in the lungs appeared to occur in two waves. The first very early wave was apparent before dividing T cells could be detected in the lymph node (LN) and coincided with neutrophil influx. The second wave of divided T cells accumulating in lung followed the appearance of these cells in LN and coincided with peak eosinophilia. Furthermore, accumulation of antigen-specific T cells in the draining LN and lung tissue, together with accompanying pathology, was reduced by intervention with the sphingosine 1-phosphate receptor agonist FTY720 2 days after challenge. These findings provide greater insight into the timing and location of antigen-specific T cell division in airways inflammation, indicate that distinct phases and locations of antigen presentation may be associated with different aspects of pathology and that therapeutics targeted against leukocyte migration may be useful in these conditions.
Collapse
Affiliation(s)
- S Hutchison
- Centre for Biophotonics, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK.
| | | | | | | | | | | | | | | |
Collapse
|
86
|
Masopust D. Developing an HIV cytotoxic T-lymphocyte vaccine: issues of CD8 T-cell quantity, quality and location. J Intern Med 2009; 265:125-37. [PMID: 19093965 DOI: 10.1111/j.1365-2796.2008.02054.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Issues of quantity, quality and location impact the ability of CD8 T cells to mediate protection from infection. These issues are considered in light of human immunodeficiency virus (HIV)/simian immunodeficiency virus (SIV) vaccination. Methods are reviewed that result in 100- to 1000-fold higher frequencies of vaccine-specific memory CD8 T cells than that achieved by current HIV/SIV vaccine approaches. Data demonstrating that location within mucosal tissues has a direct impact on memory CD8 T-cell function are discussed. Arguments are made that establishing memory CD8 T cells within mucosal sites of transmission, a priori to natural infection, may be essential for conferring optimal and rapid protection. Lastly, it is proposed that heterologous prime-boost vaccination with recombinant live replicating vectors, which has the potential to induce tremendous numbers of cytolytic memory CD8 T cells within mucosal tissues, would provide a far more stringent test of the hypothesis that memory CD8 T cells could, in principal, form the basis for a preventative HIV vaccine.
Collapse
Affiliation(s)
- D Masopust
- Department of Microbiology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
87
|
Khanna KM, Aguila CC, Redman JM, Suarez-Ramirez JE, Lefrançois L, Cauley LS. In situ imaging reveals different responses by naïve and memory CD8 T cells to late antigen presentation by lymph node DC after influenza virus infection. Eur J Immunol 2008; 38:3304-15. [PMID: 19009527 PMCID: PMC2662394 DOI: 10.1002/eji.200838602] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Pulmonary influenza infection causes prolonged lymph node hypertrophy while processed viral antigens continue to be presented to virus-specific CD8 T cells. We show that naïve, but not central/memory, nucleoprotein (NP)-specific CD8 T cells recognized antigen-bearing CD11b(+) DC in the draining lymph nodes more than 30 days after infection. After these late transfers, the naïve CD8 T cells underwent an abortive proliferative response in the mediastinal lymph node (MLN), where large clusters of partially activated cells remained in the paracortex until at least a week after transfer. A majority of the endogenous NP-specific CD8 T cells that were in the MLN between 30 and 50 days after infection also showed signs of a continuing response to antigen stimulation. A high frequency of endogenous NP-specific CD8 T cells in the MLN indicates that late antigen presentation may help shape the epitope dominance hierarchy during reinfection.
Collapse
Affiliation(s)
- Kamal M Khanna
- Department of Immunology, University of Connecticut, Farmington, CT 06032-1319, USA
| | | | | | | | | | | |
Collapse
|
88
|
|
89
|
Jenkins MR, Mintern J, La Gruta NL, Kedzierska K, Doherty PC, Turner SJ. Cell cycle-related acquisition of cytotoxic mediators defines the progressive differentiation to effector status for virus-specific CD8+ T cells. THE JOURNAL OF IMMUNOLOGY 2008; 181:3818-22. [PMID: 18768835 DOI: 10.4049/jimmunol.181.6.3818] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although analysis of virus-specific CTL function at the peak of infection suggests that granzyme (grz) and perforin (pfp) gene expression is not coregulated, early differentiation events leading to acquisition of function are poorly understood. Using a combination of CFSE dilutions and single-cell RT-PCR, effector gene expression was determined early after CTL activation. There were low levels of pfp and grz expression at division 3, with increased expression by divisions 6-8. The increase in effector mRNA expression with division correlated with increasing ex vivo cytotoxicity. Of the mRNA transcripts detected at division 3, there was an increased frequency of grzB and grzK (compared with grzA or pfp), and this pattern was also observed at later divisions. The prevalence of OT-I CTL expressing grz/pfp mRNA was equivalent for the divided CD62L(high) and CD62L(low) sets, but the concentrations of grzB protein, levels of CTL activity, and the absolute amounts of grzB transcript were substantially greater for the CD62L(low) population. Thus, while effector gene expression can be acquired early, maturation of cytotoxic capacity requires extended differentiation.
Collapse
Affiliation(s)
- Misty R Jenkins
- Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
90
|
Palendira U, Chinn R, Raza W, Piper K, Pratt G, Machado L, Bell A, Khan N, Hislop AD, Steyn R, Rickinson AB, Buckley CD, Moss P. Selective accumulation of virus-specific CD8+ T cells with unique homing phenotype within the human bone marrow. Blood 2008; 112:3293-302. [PMID: 18635810 DOI: 10.1182/blood-2008-02-138040] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The bone marrow plays a unique role within the immune system. We compared the phenotype and function of virus-specific CD8(+) T cells from matched samples of human peripheral blood and bone marrow. Analysis of virus-specific memory CD8(+) T cells showed widely divergent partition of antigen-specific populations between blood and bone marrow. T cells specific for Epstein-Barr virus (EBV) lytic antigens were enriched 3-fold in marrow compared with blood, whereas the response to EBV latent epitopes was equivalent between the 2 compartments. No difference in EBV viral load or expression of the EBV lytic protein was observed between blood and bone marrow. In direct contrast, although cytomegalo-virus (CMV)-specific T cells were the largest virus-specific population within peripheral blood, they were reduced by 60% within marrow. Bone marrow T cells were found to exhibit a unique CCR5(+)CXCR6(+)CXCR3(-) homing phenotype which has not been observed on T cells from other secondary lymphoid organs or peripheral organs. Expression of CCR5 and CXCR6 was higher on EBV-specific T cells within peripheral blood compared with CMV-specific populations. These observations identify a novel bone marrow homing phenotype for CD8(+) memory T cells, which necessitates a reevaluation of the magnitude of antigen-specific populations within the lymphoid system.
Collapse
Affiliation(s)
- Umaimainthan Palendira
- Cancer Research UK Institute for Cancer Studies, University of Birmingham, Birmingham, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Beverley PCL, Tchilian EZ. Lessons for tuberculosis vaccines from respiratory virus infection. Expert Rev Vaccines 2008; 7:1165-72. [PMID: 18844591 DOI: 10.1586/14760584.7.8.1165] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
There is a worldwide epidemic of increasingly drug-resistant TB. Bacillus Calmette-Guérin vaccination provides partial protection against disseminated disease in infants but poor protection against later pulmonary TB. Cell-mediated protection against respiratory virus infections requires the presence of T cells in lung tissues, and the most effective prime-boost immunizations for Mycobacterium tuberculosis also induce lung-resident lymphocytes. These observations need to be taken into account when designing future vaccines against M. tuberculosis.
Collapse
Affiliation(s)
- Peter Charles Leonard Beverley
- The University of Oxford, Nuffield Department of Clinical Medicine, Edward Jenner Institute for Vaccine Research, Compton, Berkshire, RG20 7NN, UK.
| | | |
Collapse
|
92
|
Verhoeven D, Teijaro JR, Farber DL. Heterogeneous memory T cells in antiviral immunity and immunopathology. Viral Immunol 2008; 21:99-113. [PMID: 18476772 DOI: 10.1089/vim.2008.0002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Memory T cells are generated following an initial viral infection, and have the potential for mediating robust protective immunity to viral re-challenge due to their rapid and enhanced functional responses. In recent years, it has become clear that the memory T cell response to most viruses is remarkably diverse in phenotype, function, and tissue distribution, and can undergo dynamic changes during its long-term maintenance in vivo. However, the role of this variegation and compartmentalizationof memory T cells in protective immunity to viruses remains unclear. In this review,we discuss the diverse features of memory T cells that can delineate different subsets, the characteristics of memory T cells thus far identified to promote protective immune responses, and how the heterogeneous nature of memory T cells may also promote immunopathology during antiviral responses. We propose that given the profound heterogeneity of memory T cells, regulation of memory T cells during secondary responses could focus the response to participation of specific subsets,and/or inhibit memory T-cell subsets and functions that can lead to immunopathology.
Collapse
Affiliation(s)
- David Verhoeven
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | |
Collapse
|
93
|
Kohlmeier JE, Miller SC, Smith J, Lu B, Gerard C, Cookenham T, Roberts AD, Woodland DL. The chemokine receptor CCR5 plays a key role in the early memory CD8+ T cell response to respiratory virus infections. Immunity 2008; 29:101-13. [PMID: 18617426 PMCID: PMC2519120 DOI: 10.1016/j.immuni.2008.05.011] [Citation(s) in RCA: 212] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Revised: 04/24/2008] [Accepted: 05/01/2008] [Indexed: 11/18/2022]
Abstract
Innate recognition of invading pathogens in peripheral tissues results in the recruitment of circulating memory CD8(+) T cells to sites of localized inflammation during the early phase of a recall response. However, the mechanisms that control the rapid recruitment of these cells to peripheral sites are poorly understood, particularly in relation to influenza and parainfluenza infections of the respiratory tract. In this study, we demonstrate a crucial role for C-C chemokine receptor 5 (CCR5) in the accelerated recruitment of memory CD8(+) T cells to the lung airways during virus challenge. Most importantly, CCR5 deficiency resulted in decreased recruitment of memory T cells expressing key effector molecules and impaired control of virus replication during the initial stages of a secondary response. These data highlight the critical importance of early memory T cell recruitment for the efficacy of cellular immunity in the lung.
Collapse
|
94
|
Abstract
The orchestrated movement of cells of the immune system is essential to generation of productive responses leading to protective memory development. Recent advances have allowed the direct microscopic visualization of lymphocyte and antigen-presenting cell migration and interaction during immune response initiation and progression. These studies have defined important characteristics of the microanatomy of lymphocyte movement, particularly in the lymph node. Moreover, the ability to track endogenous antigen-specific T cells has revealed a coordinated pathway of CD8 T cell movement in the spleen following primary and secondary infection. As a consequence, the local anatomy of secondary lymphoid tissues during infection has emerged as a critical regulator of immunity. While some of the factors responsible for the migratory cues instructing immune cell movement have been identified, much remains to be learned. Here, we provide a brief overview of studies examining CD8 T cell localization during the immune response to infection in the context of our current understanding of immune system structure.
Collapse
Affiliation(s)
- Kamal M Khanna
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030-1319, USA
| | | |
Collapse
|
95
|
Silencing of T lymphocytes by antigen-driven programmed death in recombinant adeno-associated virus vector-mediated gene therapy. Blood 2008; 113:538-45. [PMID: 18566327 DOI: 10.1182/blood-2008-01-131375] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Recombinant adeno-associated virus (rAAV) vectors are considered promising for human gene replacement because they facilitate stable expression of therapeutic proteins in transduced tissues. Whether the success of gene therapy will be influenced by cellular immune responses targeting transgene-encoded proteins that are potentially immunogenic is unknown. Here we characterized CD8(+) T-cell activity against beta-galactosidase and enhanced green fluorescent protein, model antigens containing major histocompatibility complex (MHC) class I epitopes that are constitutively produced in murine skeletal muscle after rAAV vector transduction. Antigen-specific CD8(+) T cells were detected in the spleen and liver of mice within 7 days of muscle transduction. CD8(+) T-cell frequencies in these organs were stable, and effector functions were intact for months despite ongoing antigen production in muscle. CD8(+) T cells also infiltrated transduced muscle, where frequencies were at least 5-fold higher than in untransduced spleen and liver. Significantly, the majority of antigen-specific CD8(+) T cells in vector-transduced muscle were not functional. Loss of function in the muscle was associated with programmed death of the effector cells. Stable gene expression therefore depended on selective death of CD8(+) T cells at the site of antigen production, an effective mechanism for subverting immunity that is also potentially reversible.
Collapse
|
96
|
Hardenberg G, van der Sluijs K, van der Poll T, Medema JP. APRIL affects antibody responses and early leukocyte infiltration, but not influenza A viral control. Mol Immunol 2008; 45:3050-8. [DOI: 10.1016/j.molimm.2008.03.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2008] [Revised: 03/20/2008] [Accepted: 03/24/2008] [Indexed: 12/12/2022]
|
97
|
Abstract
The ability to develop and sustain populations of memory T cells after infection or immunization is a hallmark of the adaptive immune response and a basis for protective vaccination against infectious disease. Technical advances that allow direct ex vivo identification and characterization of antigen-specific CD8+ T cells at various stages of the response to infection or vaccination in mouse models have fuelled efforts to characterize the factors that control memory CD8+ T-cell generation. Here, we dissect the input signals that shape the characteristics of the memory CD8+ T-cell response and discuss how manipulation of these signals has the potential to reshape CD8+ T-cell memory and improve the efficacy of vaccination.
Collapse
|
98
|
Broad dispersion and lung localization of virus-specific memory B cells induced by influenza pneumonia. Proc Natl Acad Sci U S A 2008; 105:3485-90. [PMID: 18299574 DOI: 10.1073/pnas.0800003105] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Although memory B cells (B(Mem)) contribute significantly to resistance to infection, B(Mem) population characteristics that may relate to protective efficacy have received little attention. Here, we report a comprehensive quantitative analysis of virus-specific IgG and IgA B(Mem) dispersion after transient influenza pneumonia in mice. From early in the response, B(Mem) circulated continuously and dispersed widely to secondary lymphoid tissues. However, a complicated picture emerged with B(Mem) frequency differences between secondary lymphoid tissues indicating an influence of local tissue factors on trafficking. B(Mem) numbers increased and stabilized at tissue-specific frequencies without contraction of the B(Mem) pool during the period of analysis. The lung was notable as a nonsecondary lymphoid tissue where a rapid influx of IgG and IgA B(Mem) established relatively high frequencies that were maintained long term. Our findings provide insights into the pattern of B(Mem) dispersion, and emphasize the lung as a complex repository of immune memory after local infection.
Collapse
|
99
|
Qu D, Lanier G, Yuan ZH, Wen YM, Howard CR, Ahmed R. Localization of CD8+ cells specific for hepatitis B virus surface protein in the liver of immunized mice. J Med Virol 2008; 80:225-32. [PMID: 18098130 DOI: 10.1002/jmv.21039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
DNA plasmids are potent inducers of long-lasting antigen-specific CTL responses. Little is known about the distribution of antigen-specific CD8+ T cells in the lymphoid tissue and the non-lymphoid tissue after DNA immunization. HBsAg-specific CD8+ T cells in peripheral blood mononuclear cells, spleen, lymph nodes, and the liver of Balb/c mice have been quantified after injection with a DNA plasmid expressing the major S protein of hepatitis B virus (HBV). The kinetics of CD8+ T-cell responses in the circulation were measured after priming and boosting, showing that antigen-specific CD8+ T cells undergo first expansion and then decline to a sustainable level in the circulation, although the frequencies of HBsAg-specific CD8+ T cells in the circulation were lower than for the spleen. The greater frequencies of HBsAg-specific CD8+ T cells were found in the liver, whereas the largest numbers of antigen-specific CD8+ T cells were found in the spleen. By day 100 after priming, HBsAg-specific CD8+ T cells were still detected in the circulation, the spleen and the liver. After boosting with the same plasmid DNA immunogen, HBsAg-specific CD8+ T cells proliferated quickly and vigorously. By 150 days after boosting, HBsAg-specific memory CD8+ T cells were sustained at higher levels than those recorded after the first, primary injection, both in the spleen and the liver: anti-HBs antibody-secreting plasma cells persisted in the bone marrow and in the spleen, consistent with the detection of anti-HBs antibodies detected in the blood. These findings indicate that DNA immunization has considerable potential for inducing specific T cell responses in the liver and offers a strategy for the development of post-exposure immunotherapy against persistent hepatitis B infections.
Collapse
Affiliation(s)
- Di Qu
- Department of Medical Molecular Virology, Institutes of Bio-medical Sciences, Shanghai Medical College of Fudan University, Shanghai, People's Republic of China
| | | | | | | | | | | |
Collapse
|
100
|
Kogan AN, von Andrian UH. Lymphocyte Trafficking. Microcirculation 2008. [DOI: 10.1016/b978-0-12-374530-9.00012-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|