101
|
Lukens JR, Dolina JS, Kim TS, Tacke RS, Hahn YS. Liver is able to activate naïve CD8+ T cells with dysfunctional anti-viral activity in the murine system. PLoS One 2009; 4:e7619. [PMID: 19876399 PMCID: PMC2764869 DOI: 10.1371/journal.pone.0007619] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2009] [Accepted: 10/05/2009] [Indexed: 11/18/2022] Open
Abstract
The liver possesses distinct tolerogenic properties because of continuous exposure to bacterial constituents and nonpathogenic food antigen. The central immune mediators required for the generation of effective immune responses in the liver environment have not been fully elucidated. In this report, we demonstrate that the liver can indeed support effector CD8+ T cells during adenovirus infection when the T cells are primed in secondary lymphoid tissues. In contrast, when viral antigen is delivered predominantly to the liver via intravenous (IV) adenovirus infection, intrahepatic CD8+ T cells are significantly impaired in their ability to produce inflammatory cytokines and lyse target cells. Additionally, intrahepatic CD8+ T cells generated during IV adenovirus infection express elevated levels of PD-1. Notably, lower doses of adenovirus infection do not rescue the impaired effector function of intrahepatic CD8+ T cell responses. Instead, intrahepatic antigen recognition limits the generation of potent anti-viral responses at both priming and effector stages of the CD8+ T cell response and accounts for the dysfunctional CD8+ T cell response observed during IV adenovirus infection. These results also implicate that manipulation of antigen delivery will facilitate the design of improved vaccination strategies to persistent viral infection.
Collapse
Affiliation(s)
- John R. Lukens
- Beirne Carter Center for Immunology Research, Department of Microbiology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Joseph S. Dolina
- Beirne Carter Center for Immunology Research, Department of Microbiology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Taeg S. Kim
- Beirne Carter Center for Immunology Research, Department of Microbiology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Robert S. Tacke
- Beirne Carter Center for Immunology Research, Department of Microbiology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Young S. Hahn
- Beirne Carter Center for Immunology Research, Department of Microbiology, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
102
|
Zhao J, Zhao J, Van Rooijen N, Perlman S. Evasion by stealth: inefficient immune activation underlies poor T cell response and severe disease in SARS-CoV-infected mice. PLoS Pathog 2009; 5:e1000636. [PMID: 19851468 PMCID: PMC2762542 DOI: 10.1371/journal.ppat.1000636] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Accepted: 09/25/2009] [Indexed: 02/04/2023] Open
Abstract
Severe Acute Respiratory Syndrome caused substantial morbidity and mortality during the 2002-2003 epidemic. Many of the features of the human disease are duplicated in BALB/c mice infected with a mouse-adapted version of the virus (MA15), which develop respiratory disease with high morbidity and mortality. Here, we show that severe disease is correlated with slow kinetics of virus clearance and delayed activation and transit of respiratory dendritic cells (rDC) to the draining lymph nodes (DLN) with a consequent deficient virus-specific T cell response. All of these defects are corrected when mice are treated with liposomes containing clodronate, which deplete alveolar macrophages (AM). Inhibitory AMs are believed to prevent the development of immune responses to environmental antigens and allergic responses by interacting with lung dendritic cells and T cells. The inhibitory effects of AM can also be nullified if mice or AMs are pretreated with poly I:C, which directly activate AMs and rDCs through toll-like receptors 3 (TLR3). Further, adoptive transfer of activated but not resting bone marrow-derived dendritic cells (BMDC) protect mice from lethal MA15 infection. These results may be relevant for SARS in humans, which is also characterized by prolonged virus persistence and delayed development of a SARS-CoV-specific immune response in individuals with severe disease.
Collapse
Affiliation(s)
- Jincun Zhao
- Department of Microbiology, University of Iowa, Iowa City, Iowa, United States of America
| | - Jingxian Zhao
- Department of Microbiology, University of Iowa, Iowa City, Iowa, United States of America
- Institute for Tissue Transplantation and Immunology, Jinan University, Guangzhou, China
| | - Nico Van Rooijen
- Department of Molecular Cell Biology, Vrije Universiteit Medisch Centrum, Amsterdam, The Netherlands
| | - Stanley Perlman
- Department of Microbiology, University of Iowa, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
103
|
Hammitt LL, Bartlett JP, Li S, Rahkola J, Lang N, Janoff EN, Levin MJ, Weinberg A. Kinetics of viral shedding and immune responses in adults following administration of cold-adapted influenza vaccine. Vaccine 2009; 27:7359-66. [PMID: 19800447 DOI: 10.1016/j.vaccine.2009.09.041] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Revised: 09/03/2009] [Accepted: 09/13/2009] [Indexed: 11/30/2022]
Abstract
The optimal type and timing of specimens to study the immune responses to cold-adapted influenza vaccine (CAIV) and shedding of vaccine virus are not well established. Healthy adults were vaccinated with CAIV (n=10) or trivalent influenza vaccine (TIV) (n=5). Shedding of vaccine strain influenza B was detected by culture in 6 of 10 CAIV recipients; influenza A was also detected in one of these subjects. Viral shedding by quantitative RT-PCR was detected in 9 of 10 subjects. We detected a > or = 2-fold increase in influenza-specific IgA in nasal wash in 80-100% of CAIV recipients following vaccination, but specific IgG increased in neither nasal wash nor saliva. Recipients of TIV had significant increases in specific serum IgG antibodies. Recipients of both CAIV and TIV had significant increases in IFNgamma-secreting peripheral blood mononuclear cells (PBMCs). PBMCs from subjects receiving CAIV showed a higher proportion of functional, tissue-tropic T-cells (CD4+CD69+CD18+MIP1alpha+) specific for homotypic and heterosubtypic strains of influenza by flow cytometry.
Collapse
Affiliation(s)
- Laura L Hammitt
- Section of Infectious Diseases, Department of Pediatrics, University of Colorado Denver, Denver, CO, United States.
| | | | | | | | | | | | | | | |
Collapse
|
104
|
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]
|
105
|
McGill J, Heusel JW, Legge KL. Innate immune control and regulation of influenza virus infections. J Leukoc Biol 2009; 86:803-12. [PMID: 19643736 DOI: 10.1189/jlb.0509368] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Adaptive immune responses are critical for the control and clearance of influenza A virus (IAV) infection. However, in recent years, it has become increasingly apparent that innate immune cells, including natural killer cells, alveolar macrophages (aMphi), and dendritic cells (DC) are essential following IAV infection in the direct control of viral replication or in the induction and regulation of virus-specific adaptive immune responses. This review will discuss the role of these innate immune cells following IAV infection, with a particular focus on DC and their ability to induce and regulate the adaptive IAV-specific immune response.
Collapse
Affiliation(s)
- Jodi McGill
- Department of Pathology and Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | |
Collapse
|
106
|
Antigen display, T-cell activation, and immune evasion during acute and chronic ehrlichiosis. Infect Immun 2009; 77:4643-53. [PMID: 19635826 DOI: 10.1128/iai.01433-08] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
How spatial and temporal changes in major histocompatibility complex/peptide antigen presentation to CD4 T cells regulate CD4 T-cell responses during intracellular bacterial infections is relatively unexplored. We have shown that immunization with an ehrlichial outer membrane protein, OMP-19, protects mice against fatal ehrlichial challenge infection, and we identified a CD4 T-cell epitope (IA(b)/OMP-19(107-122)) that elicited CD4 T cells following either immunization or infection. Here, we have used an IA(b)/OMP-19(107-122)-specific T-cell line to monitor antigen display ex vivo during acute and chronic infection with Ehrlichia muris, a bacterium that establishes persistent infection in C57BL/6 mice. The display of IA(b)/OMP-19(107-122) by host antigen-presenting cells was detected by measuring intracellular gamma interferon (IFN-gamma) production by the T-cell line. After intravenous infection, antigen presentation was detected in the spleen, peritoneal exudate cells, and lymph nodes, although the kinetics of antigen display differed among the tissues. Antigen presentation and bacterial colonization were closely linked in each anatomical location, and there was a direct relationship between antigen display and CD4 T-cell effector function. Spleen and lymph node dendritic cells (DCs) were efficient presenters of IA(b)/OMP-19(107-122), demonstrating that DCs play an important role in ehrlichial infection and immunity. Chronic infection and antigen presentation occurred within the peritoneal cavity, even in the presence of highly activated CD4 T cells. These data indicated that the ehrlichiae maintain chronic infection not by inhibiting antigen presentation or T-cell activation but, in part, by avoiding signals mediated by activated T cells.
Collapse
|
107
|
Moffat JM, Gebhardt T, Doherty PC, Turner SJ, Mintern JD. Granzyme A expression reveals distinct cytolytic CTL subsets following influenza A virus infection. Eur J Immunol 2009; 39:1203-10. [PMID: 19404988 DOI: 10.1002/eji.200839183] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
CTL mediate anti-viral immunity via targeted exocytosis of cytolytic granules containing perforin and members of the granzyme (grz) serine protease family. Here, we provide the first analysis of grzA protein expression by murine anti-viral CTL. During the progression of influenza A virus infection, CTL expressed two divergent cytolytic phenotypes: grzA(-)B(+) and grzA(+)B(+). CTL lacked grzA expression during the initial rounds of antigen-driven division. High levels of grzA were expressed by influenza-specific CTL early post infection (day 6), particularly in tissues associated with the infected respiratory tract (bronchoalveolar lavage, lung). Following resolution of influenza infection, a small population of memory CTL expressed grzA. Interestingly, individual influenza A virus-derived epitope-specific CTL expressed different levels of grzA. The grzA expression hierarchy was determined to be K(b)PB1(703)=D(b)F2(62)=K(b)NS2(114)>D(b)NP(366)=D(b)PA(224) and inversely correlated with CTL magnitude. Therefore following influenza infection, a CTL cytolytic hierarchy was established relating to the different profiles of antigen expression and relative immunodominance. Analysis of CTL grzA expression during influenza virus immunity has enabled a more detailed insight into the cytolytic mechanisms of virus elimination.
Collapse
Affiliation(s)
- Jessica M Moffat
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, Victoria, Australia
| | | | | | | | | |
Collapse
|
108
|
Flaño E, Jewell NA, Durbin RK, Durbin JE. Methods used to study respiratory virus infection. CURRENT PROTOCOLS IN CELL BIOLOGY 2009; Chapter 26:Unit 26.3. [PMID: 19499505 PMCID: PMC2753376 DOI: 10.1002/0471143030.cb2603s43] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
This unit describes protocols for infecting the mouse respiratory tract, and assaying virus replication and host response in the lung. Respiratory infections are the leading cause of acute illness worldwide, affecting mostly infants and children in developing countries. The purpose of this unit is to provide a basic strategy and protocols to study the pathogenesis and immunology of respiratory virus infection using the mouse as an animal model. The procedures include: (1) basic techniques for mouse infection, tissue sampling, and preservation, (2) determination of viral titers, isolation and analysis of lymphocytes and dendritic cells using flow-cytometry, and (3) lung histology, immunohistochemistry, and in situ hybridization.
Collapse
Affiliation(s)
- Emilio Flaño
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital, Ohio State University, Columbus, Ohio, USA
| | | | | | | |
Collapse
|
109
|
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
|
110
|
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
|
111
|
Gelman AE, Li W, Richardson SB, Zinselmeyer BH, Lai J, Okazaki M, Kornfeld CG, Kreisel FH, Sugimoto S, Tietjens JR, Dempster J, Patterson GA, Krupnick AS, Miller MJ, Kreisel D. Cutting edge: Acute lung allograft rejection is independent of secondary lymphoid organs. THE JOURNAL OF IMMUNOLOGY 2009; 182:3969-73. [PMID: 19299693 DOI: 10.4049/jimmunol.0803514] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
It is the prevailing view that adaptive immune responses are initiated in secondary lymphoid organs. Studies using alymphoplastic mice have shown that secondary lymphoid organs are essential to initiate allograft rejection of skin, heart, and small bowel. The high immunogenicity of lungs is well recognized and allograft rejection remains a major contributing factor to poor outcomes after lung transplantation. We show in this study that alloreactive T cells are initially primed within lung allografts and not in secondary lymphoid organs following transplantation. In contrast to other organs, lungs are acutely rejected in the absence of secondary lymphoid organs. Two-photon microscopy revealed that recipient T cells cluster predominantly around lung-resident, donor-derived CD11c(+) cells early after engraftment. These findings demonstrate for the first time that alloimmune responses following lung transplantation are initiated in the graft itself and therefore identify a novel, potentially clinically relevant mechanism of lung allograft rejection.
Collapse
Affiliation(s)
- Andrew E Gelman
- Department of Surgery, Washington University, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
112
|
Sun J, Madan R, Karp CL, Braciale TJ. Effector T cells control lung inflammation during acute influenza virus infection by producing IL-10. Nat Med 2009; 15:277-84. [PMID: 19234462 PMCID: PMC2693210 DOI: 10.1038/nm.1929] [Citation(s) in RCA: 490] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2008] [Accepted: 01/13/2009] [Indexed: 12/14/2022]
Abstract
Activated antigen-specific T cells produce a variety of effector molecules for clearing infection but also contribute to inflammation and tissue injury. Here we report an anti-inflammatory property of antiviral CD8+ and CD4+ effector T cells (T(eff) cells) in the infected periphery during acute virus infection. We find that, during acute influenza infection, interleukin-10 (IL-10) is produced in the infected lungs in large amounts--exclusively by infiltrating virus-specific T(eff) cells, with CD8+ T(eff) cells contributing a larger fraction of the IL-10 produced. These T(eff) cells in the periphery simultaneously produce IL-10 and proinflammatory cytokines and express lineage markers characteristic of conventional T helper type 1 or T cytotoxic type 1 cells. Notably, blocking the action of the T(eff) cell-derived IL-10 results in enhanced pulmonary inflammation and lethal injury. Our results show that antiviral T(eff) cells exert regulatory functions--that is, they fine-tune the extent of lung inflammation and injury associated with influenza infection by producing an anti-inflammatory cytokine. We discuss the potential implications of these findings for infection with highly pathogenic influenza viruses.
Collapse
Affiliation(s)
- Jie Sun
- Beirne B. Carter Center for Immunology Research, University of Virginia, 409 Lane Road, Charlottesville, VA 22908
| | - Rajat Madan
- Division of Molecular Immunology, Cincinnati Children’s Hospital Research Foundation, and the University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, Ohio 45229, USA
| | - Christopher L. Karp
- Division of Molecular Immunology, Cincinnati Children’s Hospital Research Foundation, and the University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, Ohio 45229, USA
| | - Thomas J. Braciale
- Beirne B. Carter Center for Immunology Research, University of Virginia, 409 Lane Road, Charlottesville, VA 22908
- Departments of Microbiology and Pathology, University of Virginia, 409 Lane Road, Charlottesville, VA 22908
| |
Collapse
|
113
|
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
|
114
|
Yates A. Modelling pathways of CD8+ T-cell differentiation. Eur J Immunol 2009; 39:47-9. [PMID: 19130546 DOI: 10.1002/eji.200839063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
In an immune response to infection, naïve T lymphocytes proliferate and give rise to a heterogeneous population of effector and memory cells. How is this diversity generated, and how can it be manipulated? Answering these questions requires an understanding of the lineage relationships between different effector and memory-cell subsets, but these relationships remain to be identified definitively. In this issue of the European Journal of Immunology, a study moves us closer to this goal by combining a mathematical model and data from influenza infections in mice to support the hypothesis that CD8(+) T-cell differentiation is strongly coupled to cell division.
Collapse
Affiliation(s)
- Andrew Yates
- Department of Biology, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
115
|
Kim TS, Braciale TJ. Respiratory dendritic cell subsets differ in their capacity to support the induction of virus-specific cytotoxic CD8+ T cell responses. PLoS One 2009; 4:e4204. [PMID: 19145246 PMCID: PMC2615220 DOI: 10.1371/journal.pone.0004204] [Citation(s) in RCA: 225] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Accepted: 11/25/2008] [Indexed: 02/06/2023] Open
Abstract
Dendritic cells located at the body surfaces, e.g. skin, respiratory and gastrointestinal tract, play an essential role in the induction of adaptive immune responses to pathogens and inert antigens present at these surfaces. In the respiratory tract, multiple subsets of dendritic cells (RDC) have been identified in both the normal and inflamed lungs. While the importance of RDC in antigen transport from the inflamed or infected respiratory tract to the lymph nodes draining this site is well recognized, the contribution of individual RDC subsets to this process and the precise role of migrant RDC within the lymph nodes in antigen presentation to T cells is not clear. In this report, we demonstrate that two distinct subsets of migrant RDC--exhibiting the CD103(+) and CD11b(hi) phenotype, respectively--are the primary DC presenting antigen to naïve CD4(+) and CD8(+) T lymphocytes in the draining nodes in response to respiratory influenza virus infection. Furthermore, the migrant CD103(+) RDC subset preferentially drives efficient proliferation and differentiation of naive CD8(+) T cells responding to infection into effector cells, and only the CD103(+) RDC subset can present to naïve CD8(+) T cells non-infectious viral vaccine introduced into the respiratory tract. These results identify CD103(+) and CD11b(hi) RDC as critical regulators of the adaptive immune response to respiratory tract infection and potential targets in the design of mucosal vaccines.
Collapse
Affiliation(s)
- Taeg S. Kim
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, United States of America
| | - Thomas J. Braciale
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, United States of America
- Departments of Microbiology and Pathology, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
116
|
Schlub TE, Venturi V, Kedzierska K, Wellard C, Doherty PC, Turner SJ, Ribeiro RM, Hodgkin PD, Davenport MP. Division-linked differentiation can account for CD8+ T-cell phenotype in vivo. Eur J Immunol 2009; 39:67-77. [DOI: 10.1002/eji.200838554] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
117
|
Ingulli E, Funatake C, Jacovetty EL, Zanetti M. Cutting Edge: Antigen Presentation to CD8 T Cells after Influenza A Virus Infection. THE JOURNAL OF IMMUNOLOGY 2008; 182:29-33. [DOI: 10.4049/jimmunol.182.1.29] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
118
|
Droebner K, Haasbach E, Fuchs C, Weinzierl AO, Stevanovic S, Büttner M, Planz O. Antibodies and CD4+ T-cells mediate cross-protection against H5N1 influenza virus infection in mice after vaccination with a low pathogenic H5N2 strain. Vaccine 2008; 26:6965-74. [DOI: 10.1016/j.vaccine.2008.09.051] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2008] [Revised: 09/10/2008] [Accepted: 09/17/2008] [Indexed: 10/21/2022]
|
119
|
Robila V, Ostankovitch M, Altrich-VanLith ML, Theos AC, Drover S, Marks MS, Restifo N, Engelhard VH. MHC class II presentation of gp100 epitopes in melanoma cells requires the function of conventional endosomes and is influenced by melanosomes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:7843-52. [PMID: 19017974 PMCID: PMC2659719 DOI: 10.4049/jimmunol.181.11.7843] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Many human solid tumors express MHC class II (MHC-II) molecules, and proteins normally localized to melanosomes give rise to MHC-II-restricted epitopes in melanoma. However, the pathways by which this response occurs have not been defined. We analyzed the processing of one such epitope, gp100(44-59), derived from gp100/Pmel17. In melanomas that have down-regulated components of the melanosomal pathway, but constitutively express HLA-DR*0401, the majority of gp100 is sorted to LAMP-1(high)/MHC-II(+) late endosomes. Using mutant gp100 molecules with altered intracellular trafficking, we demonstrate that endosomal localization is necessary for gp100(44-59) presentation. By depletion of the AP-2 adaptor protein using small interfering RNA, we demonstrate that gp100 protein internalized from the plasma membrane to such endosomes is a major source for gp100(44-59) epitope production. The gp100 trapped in early endosomes gives rise to epitopes that are indistinguishable from those produced in late endosomes but their production is less sensitive to inhibition of lysosomal proteases. In melanomas containing melanosomes, gp100 is underrepresented in late endosomes, and accumulates in stage II melanosomes devoid of MHC-II molecules. The gp100(44-59) presentation is dramatically reduced, and processing occurs entirely in early endosomes or stage I melanosomes. This occurrence suggests that melanosomes are inefficient Ag-processing compartments. Thus, melanoma de-differentiation may be accompanied by increased presentation of MHC-II restricted epitopes from gp100 and other melanosome-localized proteins, leading to enhanced immune recognition.
Collapse
Affiliation(s)
- Valentina Robila
- Department of Microbiology and Carter Immunology Center, University of Virginia School of Medicine, Charlottesville VA 22908
| | - Marina Ostankovitch
- Department of Microbiology and Carter Immunology Center, University of Virginia School of Medicine, Charlottesville VA 22908
| | - Michelle L. Altrich-VanLith
- Department of Microbiology and Carter Immunology Center, University of Virginia School of Medicine, Charlottesville VA 22908
| | - Alexander C. Theos
- Dept. of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia PA 19104
| | - Sheila Drover
- Division of Biomedical Sciences, Memorial University of Newfoundland, St. John's, NF Canada A1B3V6
| | - Michael S. Marks
- Dept. of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia PA 19104
| | - Nicholas Restifo
- National Cancer Institute, National Institutes of Health Bethesda, MD 20892
| | - Victor H. Engelhard
- Department of Microbiology and Carter Immunology Center, University of Virginia School of Medicine, Charlottesville VA 22908
| |
Collapse
|
120
|
Fadel SA, Bromley SK, Medoff BD, Luster AD. CXCR3-deficiency protects influenza-infected CCR5-deficient mice from mortality. Eur J Immunol 2008; 38:3376-87. [PMID: 19039768 PMCID: PMC2749081 DOI: 10.1002/eji.200838628] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Mice lacking the chemokine receptor CCR5 are susceptible to mortality from a normally non-lethal influenza infection. Here we found that CXCR3-deficiency rescued CCR5-deficient (CCR5(-/-)) mice from influenza-induced mortality. The number of mononuclear phagocytes in the airways was transiently increased in CCR5(-/-) mice but not in CXCR3-CCR5 double-deficient mice. Antigen-specific CXCR3-CCR5 double-deficient CD8 effector cells were less efficient at entering the airways compared with WT or CCR5(-/-) CD8 effector cells. The decrease in inflammatory cell infiltrates in CXCR3-CCR5 double-deficient-infected mice correlated with a decrease in CCL2 and IFN-gamma production in the airways. Finally, CXCR3-CCR5 double-deficient mice that survived the primary viral challenge were protected from a lethal secondary challenge, indicating that T-cell-mediated protective memory was not compromised in mice lacking these chemokine receptors. In conclusion, CXCR3-deficiency attenuated the lethal cellular immune response in CCR5(-/-) influenza-infected mice without hindering viral clearance or long-term immunity.
Collapse
MESH Headings
- Animals
- Cell Movement/immunology
- Female
- Influenza A virus/immunology
- Lymphocytes/cytology
- Lymphocytes/immunology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/pathology
- Receptors, CCR5/deficiency
- Receptors, CCR5/genetics
- Receptors, CCR5/immunology
- Receptors, CCR5/metabolism
- Receptors, CXCR3/deficiency
- Receptors, CXCR3/genetics
- Receptors, CXCR3/immunology
- Receptors, CXCR3/metabolism
- Survival Rate
Collapse
Affiliation(s)
- Shaza A. Fadel
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Shannon K. Bromley
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Benjamin D. Medoff
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Pulmonary and Critical Care Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Andrew D. Luster
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
121
|
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
|
122
|
Abstract
SUMMARY Tuberculosis (TB) has plagued mankind for millennia yet is classified as an emerging infectious disease, because its prevalence in the human population continues to increase. Immunity to TB depends critically on the generation of effective CD4(+) T-cell responses. Sterile immunity has not been achieved through vaccination, although early T-cell responses are effective in controlling steady-state infection in the lungs. Although such early T-cell responses are clearly protective, the initiation of the Mycobacterium tuberculosis (Mtb) T-cell response occurs much later than is the case following other aerogenic infections. This fact suggests that there is a critical period, before the activation of the T-cell response, in which Mtb is able to establish infection. An understanding of the factors that regulate early T-cell activation should, therefore, lead to better control of the disease. This review discusses recent work that has investigated the early development of T-cell immunity following Mtb infection in the mouse.
Collapse
Affiliation(s)
- Gary M Winslow
- New York State Department of Health, Wadsworth Center, Albany, NY 12208, USA.
| | | | | | | | | |
Collapse
|
123
|
Brincks EL, Katewa A, Kucaba TA, Griffith TS, Legge KL. CD8 T cells utilize TRAIL to control influenza virus infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:4918-25. [PMID: 18802095 PMCID: PMC2610351 DOI: 10.4049/jimmunol.181.7.4918] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Elimination of influenza virus-infected cells during primary influenza virus infections is thought to be mediated by CD8(+) T cells though perforin- and FasL-mediated mechanisms. However, recent studies suggest that CD8(+) T cells can also utilize TRAIL to kill virally infected cells. Therefore, we herein examined the importance of TRAIL to influenza-specific CD8(+) T cell immunity and to the control of influenza virus infections. Our results show that TRAIL deficiency increases influenza-associated morbidity and influenza virus titers, and that these changes in disease severity are coupled to decreased influenza-specific CD8(+) T cell cytotoxicity in TRAIL(-/-) mice, a decrease that occurs despite equivalent numbers of pulmonary influenza-specific CD8(+) T cells. Furthermore, TRAIL expression occurs selectively on influenza-specific CD8(+) T cells, and high TRAIL receptor (DR5) expression occurs selectively on influenza virus-infected pulmonary epithelial cells. Finally, we show that adoptive transfer of TRAIL(+/+) but not TRAIL(-/-) CD8(+) effector T cells alters the mortality associated with lethal dose influenza virus infections. Collectively, our results suggest that TRAIL is an important component of immunity to influenza infections and that TRAIL deficiency decreases CD8(+) T cell-mediated cytotoxicity, leading to more severe influenza infections.
Collapse
Affiliation(s)
- Erik L. Brincks
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA
- Department of Urology, University of Iowa, Iowa City, IA
| | - Arna Katewa
- Department of Pathology, University of Iowa, Iowa City, IA
| | | | - Thomas S. Griffith
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA
- Department of Urology, University of Iowa, Iowa City, IA
| | - Kevin L. Legge
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA
- Department of Pathology, University of Iowa, Iowa City, IA
| |
Collapse
|
124
|
Brincks EL, Kucaba TA, Legge KL, Griffith TS. Influenza-induced expression of functional tumor necrosis factor-related apoptosis-inducing ligand on human peripheral blood mononuclear cells. Hum Immunol 2008; 69:634-46. [PMID: 18723061 PMCID: PMC2597454 DOI: 10.1016/j.humimm.2008.07.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2008] [Revised: 07/14/2008] [Accepted: 07/18/2008] [Indexed: 12/30/2022]
Abstract
The immunologic response to influenza virus infection, like many other viruses, is characterized by robust production of proinflammatory cytokines, including type I and II interferon (IFN), which induce a number of antiviral effects and are essential for priming the innate and adaptive cellular components of the immune response. Here, we demonstrate that influenza virus infection induces the expression of functional tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) on human peripheral blood mononuclear cell (PBMC) populations. Consistent with previous studies examining TRAIL upregulation, increased TRAIL expression correlated with increased type I and II IFN levels in PBMC cultures. Interestingly, dilution of these cytokines resulted in decreased expression of TRAIL. TRAIL upregulation was not dependent on active viral infection, and TRAIL was observed on NS-1-negative cells. Furthermore, influenza virus infection of lung adenocarcinoma cells (A549) resulted in increased sensitization to TRAIL-induced apoptosis compared with uninfected A549. Infected PBMC expressing TRAIL preferentially killed infected A549, but did not affect uninfected cells, and the addition of soluble TRAIL-R2:Fc blocked the lysis of infected cells, demonstrating TRAIL-dependent killing of infected cells. Collectively, these data demonstrate that TRAIL expression is induced on primary human innate and adaptive immune cells in response to cytokines produced during influenza infection and that TRAIL sensitivity is increased in influenza virus-infected cells. These data also suggest that TRAIL is a primary mechanism used by influenza-stimulated human PBMC to kill influenza-infected target cells and reinforce the importance of cytokines produced in response to TLR agonists in enhancing cellular immune effector functions.
Collapse
Affiliation(s)
- Erik L. Brincks
- Department of Urology, University of Iowa, 375 Newton Road, Iowa City, IA 52242
- Interdisciplinary Graduate Program in Immunology, University of Iowa, 375 Newton Road, Iowa City, IA 52242
| | - Tamara A. Kucaba
- Department of Urology, University of Iowa, 375 Newton Road, Iowa City, IA 52242
| | - Kevin L. Legge
- Department of Pathology, University of Iowa, 375 Newton Road, Iowa City, IA 52242
- Interdisciplinary Graduate Program in Immunology, University of Iowa, 375 Newton Road, Iowa City, IA 52242
| | - Thomas S. Griffith
- Department of Urology, University of Iowa, 375 Newton Road, Iowa City, IA 52242
- Interdisciplinary Graduate Program in Immunology, University of Iowa, 375 Newton Road, Iowa City, IA 52242
| |
Collapse
|
125
|
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
|
126
|
Brinkman CC, Sheasley-O'Neill SL, Ferguson AR, Engelhard VH. Activated CD8 T cells redistribute to antigen-free lymph nodes and exhibit effector and memory characteristics. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:1814-24. [PMID: 18641319 PMCID: PMC2591092 DOI: 10.4049/jimmunol.181.3.1814] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Exogenous dendritic cells display restricted trafficking when injected in vivo and stimulate CD8 T cell responses that are localized to a small number of lymphoid compartments. By examining these responses in the presence and absence of FTY720, a drug that causes sequestration of T cells in lymph nodes, we demonstrate that a significant fraction of divided CD8 T cells redistribute into Ag-free lymph nodes within 3 days of activation. Despite variation in the level of expression of CD62L, redistribution of these cells is CD62L-dependent. Redistributed CD8 T cells exhibit characteristics of differentiated effectors. However, when re-isolated from Ag-free lymph nodes 3 days after activation and transferred into naive mice, they persist for at least 3 wk and expand upon Ag challenge. Thus, CD8 T cells that redistribute to Ag-free lymph nodes 3 days after immunization contain memory precursors. We suggest that this redistribution process represents an important mechanism for establishment of lymph node resident central memory, and that redistribution to Ag-free nodes is an additional characteristic to be added to those that distinguish memory precursors from terminal effectors.
Collapse
Affiliation(s)
- C Colin Brinkman
- Department of Microbiology and Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | |
Collapse
|
127
|
ESAT-6-specific CD4 T cell responses to aerosol Mycobacterium tuberculosis infection are initiated in the mediastinal lymph nodes. Proc Natl Acad Sci U S A 2008; 105:10961-6. [PMID: 18667699 DOI: 10.1073/pnas.0801496105] [Citation(s) in RCA: 171] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
CD4(+) T cell responses to aerosol Mycobacterium tuberculosis (Mtb) infection are characterized by the relatively delayed appearance of effector T cells in the lungs. This delay in the adaptive response is likely critical in allowing the bacteria to establish persistent infection. Because of limitations associated with the detection of low frequencies of naïve T cells, it had not been possible to precisely determine when and where naïve antigen-specific T cells are first activated. We have addressed this problem by using early secreted antigenic target 6 (ESAT-6)-specific transgenic CD4 T cells to monitor early T cell activation in vivo. By using an adoptive transfer approach, we directly show that T cell priming to ESAT-6 occurs only after 10 days of infection, is initially restricted to the mediastinal lymph nodes, and does not involve other lymph nodes or the lungs. Primed CD4 T cells rapidly differentiated into proliferating effector cells and ultimately acquired the ability to produce IFN-gamma and TNF-alpha ex vivo. Initiation of T cell priming was enhanced by two full days depending on the magnitude of the challenge inoculum, which suggests that antigen availability is a factor limiting the early CD4 T cell response. These data define a key period in the adaptive immune response to Mtb infection.
Collapse
|
128
|
McGill J, Van Rooijen N, Legge KL. Protective influenza-specific CD8 T cell responses require interactions with dendritic cells in the lungs. ACTA ACUST UNITED AC 2008; 205:1635-46. [PMID: 18591411 PMCID: PMC2442641 DOI: 10.1084/jem.20080314] [Citation(s) in RCA: 193] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Influenza infections induce a rapid, but transient, dendritic cell (DC) migration from the lungs to the lymph nodes (LNs) that is followed by substantial recruitment of DCs into the lungs without subsequent migration to the LNs. Given that peripheral DCs are primarily thought to be involved in the initiation of adaptive immunity after migration into lymphoid tissues, what role these newly lung-recruited DCs play in influenza virus immunity is unclear. In this study, we demonstrate that loss of non-LN migratory pulmonary DC subsets increases mortality, sustains higher viral titers, and impairs pulmonary CD8 T cell responses. Reconstitution of the lungs with pulmonary plasmacytoid DCs, CD8α+ DCs, or interstitial DCs restores CD8 T cell responses in a cell contact–, major histocompatability complex I–, and influenza peptide–dependent manner. Thus, after their initial activation in the LN, protective influenza-specific CD8 T cell responses require additional antigen-dependent interactions, specifically with DCs in the lungs.
Collapse
Affiliation(s)
- Jodi McGill
- Department of Pathology, Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52241, USA
| | | | | |
Collapse
|
129
|
Meyerholz DK, Edsen-Moore M, McGill J, Coleman RA, Cook RT, Legge KL. Chronic alcohol consumption increases the severity of murine influenza virus infections. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:641-8. [PMID: 18566431 PMCID: PMC2504736 DOI: 10.4049/jimmunol.181.1.641] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Respiratory infections with both seasonal as well as potential pandemic Influenza viruses represent a significant burden on human health. Furthermore, viruses such as Influenza are increasingly recognized as important etiologic agents in community acquired pneumonia. Within the U.S. alone, approximately 12.9 million people are heavy drinkers and chronic abuse of alcohol is known to increase the risk and severity of community acquired pneumonia. Given the lack of knowledge regarding Influenza disease in this population, we determined the effects of chronic alcohol consumption on Influenza virus infection. Herein, we report that mice exposed to chronic ethanol have sharp increases in morbidity, mortality, and pulmonary virus titers relative to controls. These increases in influenza severity correspond with inhibited pulmonary influenza-specific CD8 T cell responses. Further, chronic ethanol consumption results in an enhanced pulmonary lesion severity, similar to that recently described for pandemic influenzas. Together, our results suggest that chronic alcohol consumption may increase the risk for severe influenza virus infections by altering the pulmonary inflammatory environment and CD8 T cell response.
Collapse
Affiliation(s)
| | | | - Jodi McGill
- Department of Pathology, University of Iowa, Iowa City, IA
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA
| | | | - Robert T Cook
- Department of Pathology, University of Iowa, Iowa City, IA
| | - Kevin L. Legge
- Department of Pathology, University of Iowa, Iowa City, IA
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA
| |
Collapse
|
130
|
Kushwah R, Cao H, Hu J. Characterization of pulmonary T cell response to helper-dependent adenoviral vectors following intranasal delivery. THE JOURNAL OF IMMUNOLOGY 2008; 180:4098-108. [PMID: 18322220 DOI: 10.4049/jimmunol.180.6.4098] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In spite of the extensive research in the field of gene therapy, host immune responses continue to be the major barrier in translating basic research to clinical practice. Helper-dependent adenoviral (HD-Ad) vectors show great potential for pulmonary gene therapy, but the knowledge of pulmonary immune responses toward these vectors is very limited. In this study, we show that HD-Ad vectors are potent stimulators of dendritic cell (DC) maturation, thus leading to stimulation of T cell proliferation with approximately 6% of naive CD4(+) T cells from pulmonary mediastinal lymph node responding to HD-Ad-treated DCs. In contrast to the belief that HD-Ad vectors are unable to prime adaptive immune response, we show for the first time, through in vivo pulmonary studies in mice, that HD-Ad vectors can prime CD4(+) and CD8(+) T cell responses in the lung at high and substantially low doses. This indicates cross-presentation of HD-Ad-derived epitopes by DCs to prime CD8(+) T cell responses. To assess the basis of pulmonary T cell response against HD-Ad vectors, we examined the response of conventional DCs (cDCs) and plasmacytoid DCs (pDCs) in the lung. In response to HD-Ad delivery, there is induction of maturation in both cDC and pDC subsets, but it is the cDCs, not pDCs, that migrate rapidly to draining lymph nodes within the first 2 days after vector delivery to prime adaptive immune response against these vectors. These findings have implications for development of strategies to prevent adaptive immune responses against gene therapy vectors.
Collapse
Affiliation(s)
- Rahul Kushwah
- Physiology and Experimental Medicine Research Program, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
131
|
Ag-specific type 1 CD8 effector cells enhance methotrexate-mediated antitumor responses by modulating differentiated T cell localization, activation and chemokine production in established breast cancer. Clin Immunol 2008; 128:205-18. [PMID: 18511346 DOI: 10.1016/j.clim.2008.03.518] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2007] [Revised: 03/11/2008] [Accepted: 03/26/2008] [Indexed: 01/12/2023]
Abstract
The chemotherapeutic agent methotrexate is widely used in the treatment of breast cancer. Although its mechanism-of-action has been defined, less is known about its interaction with T cell-mediated antitumor responses. Type 1 CD8 T cell-mediated immune responses (Tc1) are cytolytic, produce IFN-gamma and are associated with effective antitumor responses. Using a murine transgenic TCR tumor model, we show that single-dose treatment with methotrexate enhanced CD8-mediated type 1 antitumor responses when administered 3 days prior to Tc1 effector cell transfer. Co-treatment with methotrexate not only enhanced donor Tc1 cell accumulation and persistence at sites of primary tumor growth, but also promoted elevated levels of activated donor TIL cells. This markedly enhanced the appearance of endogenous differentiated (CD44(High)) CD8 tumor-infiltrating cells when compared to that of corresponding groups receiving either MTX or Tc1 cell transfer alone. Such cells were acutely activated as defined by co-expression of surface markers associated with TCR engagement (CD69) and T cell activation (CD25) at both early (days 1-8) and late (days 12-20) stages following treatment. Conversely, such animals showed an early decrease in CD4(+)/CD44(High)/CD25(+)/CD69(+) T cells that correlated with delays in tumor growth in vivo. Moreover, cellular response kinetics appeared to further correlate with the up-regulation of endogenous T cells producing the chemokine IP-10 in vivo. This suggested that Tc1 cell transfer, in combination with chemotherapy, can enhance antitumor responses by modulating immunoregulatory T cells involved in homeostasis and immune tolerance within the tumor environment. These studies offer insight into mechanisms that enhance T cell-based immunotherapy in cancer.
Collapse
|
132
|
Casillas S, Herrero Fernández S, Varon J. [Bird flu: what the intensivist must know]. Med Intensiva 2008; 32:183-93. [PMID: 18413124 DOI: 10.1016/s0210-5691(08)70936-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
In the last century, humankind has faced 3 major pandemics of influenza virus infections. The first one occurred in 1918 and caused a significant amount of deaths. It was also capable of crossing over species barrier and affecting mammals, and most worrisome, humans. Since then several outbreaks have been reported in the Southeast of Asia. Many patients with the flu-like illness have a severe course and the patient develops pneumonia and in some cases multiorgan failure involving liver, kidneys, brain and lungs. Since the virus lacks regulatory control of genetic division it undergoes constant mutations leading to new subtypes and, sometimes, new strains. The only drugs that have shown some protection are oseltamivir and zanamivir. It is crucial to develop effective and non-expensive vaccines to prevent the virus spread and infection not only in humans but in birds too.
Collapse
Affiliation(s)
- S Casillas
- Universidad Autónoma de Baja California, México
| | | | | |
Collapse
|
133
|
Fernandez MA, Puttur FK, Wang YM, Howden W, Alexander SI, Jones CA. T regulatory cells contribute to the attenuated primary CD8+ and CD4+ T cell responses to herpes simplex virus type 2 in neonatal mice. THE JOURNAL OF IMMUNOLOGY 2008; 180:1556-64. [PMID: 18209051 DOI: 10.4049/jimmunol.180.3.1556] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The first weeks of life are characterized by immune tolerance and increased susceptibility to intracellular pathogens. The neonatal adaptive response to HSV is attenuated compared with adult control models in humans and mice. T Regulatory cells (Tregs) control autoimmunity and excessive immune responses to infection. We therefore compared Treg responses in the draining lymph nodes (LN) of HSV-infected neonatal and adult C57BL/6 mice with the effect of Treg depletion/inactivation by anti-CD25 (PC61) treatment before infection on Ag-specific T cell effector responses at this site. There was a small, but significant increase in the frequency of CD4(+)Foxp3(+) Tregs at day 3 postinfection (p.i.) in the LN of neonatal and adult mice, compared with age-matched mock-infected controls. Depletion of Tregs before HSV infection significantly enhanced HSV-specific CD8(+) T cell cytotoxicity in vivo, cell number, activation, and granzyme B expression 4 days p.i. only in neonatal mice, and significantly enhanced CD8(+) and CD4(+) T cell IFN-gamma responses in both infected adults and neonates. Treg depletion also reduced the titer of infectious virus in the draining LN and nervous system of infected neonates on days 2 and 3 p.i. Treg suppression of the neonatal CTL response p.i. with HSV was associated with increased expression of TGF-beta in the draining LN at day 4 p.i. compared with uninfected neonates, but IL-10 was increased in infected adults alone. These experiments support the notion that the newborn primary T cell effector responses to HSV are suppressed by Tregs.
Collapse
Affiliation(s)
- Marian A Fernandez
- Centre for Perinatal Infection Research, The Children's Hospital at Westmead, Westmead, New South Wales 2145, Australia
| | | | | | | | | | | |
Collapse
|
134
|
Shen CH, Ge Q, Talay O, Eisen HN, García-Sastre A, Chen J. Loss of IL-7R and IL-15R expression is associated with disappearance of memory T cells in respiratory tract following influenza infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 180:171-8. [PMID: 18097017 PMCID: PMC2709277 DOI: 10.4049/jimmunol.180.1.171] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Following influenza virus infection, memory CD8 T cells are found in both lymphoid and nonlymphoid organs, where they exhibit striking differences in survival. We have assessed persistence, phenotype, and function of memory CD8 T cells expressing the same TCR in the airways, lung parenchyma, and spleen following influenza virus infection in mice. In contrast to memory CD8 T cells in the spleen, those residing in the airways gradually lost expression of IL-7R and IL-15R, did not respond to IL-7 and/or IL-15, and exhibited poor survival both in vivo and in vitro. Following adoptive transfer into the airways, splenic memory CD8 T cells also down-regulated IL-7R and IL-15R expression and failed to undergo homeostatic proliferation. Thus, although cytokines IL-7 and IL-15 play an essential role in memory CD8 T cell homeostasis in lymphoid organs, the levels of IL-7R and IL-15R expression likely set a threshold for the homeostatic regulation of memory CD8 T cells in the airways. These findings provide a molecular explanation for the gradual loss of airway memory CD8 T cells and heterosubtypic immunity following influenza infection.
Collapse
Affiliation(s)
- Ching-Hung Shen
- Center for Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Qing Ge
- Center for Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Oezcan Talay
- Center for Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Herman N. Eisen
- Center for Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | | | - Jianzhu Chen
- Center for Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| |
Collapse
|
135
|
Fernandez M, Evans I, Hassan E, Carbone F, Jones C. Neonatal CD8+ T cells are slow to develop into lytic effectors after HSV infectionin vivo. Eur J Immunol 2008; 38:102-13. [DOI: 10.1002/eji.200636945] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
136
|
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
|
137
|
Polakos NK, Klein I, Richter MV, Zaiss DM, Giannandrea M, Crispe IN, Topham DJ. Early intrahepatic accumulation of CD8+ T cells provides a source of effectors for nonhepatic immune responses. THE JOURNAL OF IMMUNOLOGY 2007; 179:201-10. [PMID: 17579039 DOI: 10.4049/jimmunol.179.1.201] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Interactions between the liver and CD8+ T cells can lead to tolerance, due in part to CD8+ T cell death. To test whether this was the case in an extrahepatic infection, we investigated the fate and effector capacity of intrahepatic CD8+ T cells during lung-restricted influenza infection in mice. Virus-specific T cells accumulated in livers without detectable intrahepatic presentation of viral Ags, and this accumulation was not restricted to the contraction phase, but was apparent as early as day 5. Intrahepatic influenza-specific cells were functionally similar to those recovered from the bronchioalveolar lavage, based on ex vivo cytokine production and specific target lysis. Both adoptive transfer of liver lymphocytes and orthotopic liver transplant of organs containing accumulated effector T cells revealed that activated CD8s from the liver were viable, expanded during reinfection, and generated a memory population that trafficked to lymphoid organs. Thus, intrahepatic CD8+ T cells re-enter circulation and generate functional memory, indicating that the liver does not uniformly incapacitate activated CD8+ T cells. Instead, it constitutes a substantial reservoir of usable Ag-specific effector CD8+ T cells involved in both acute and recall immune responses.
Collapse
Affiliation(s)
- Noelle K Polakos
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | | | | | | | | | | | | |
Collapse
|
138
|
Yoon H, Legge KL, Sung SSJ, Braciale TJ. Sequential activation of CD8+ T cells in the draining lymph nodes in response to pulmonary virus infection. THE JOURNAL OF IMMUNOLOGY 2007; 179:391-9. [PMID: 17579060 DOI: 10.4049/jimmunol.179.1.391] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We have used a TCR-transgenic CD8+ T cell adoptive transfer model to examine the tempo of T cell activation and proliferation in the draining lymph nodes (DLN) in response to respiratory virus infection. The T cell response in the DLN differed for mice infected with different type A influenza strains with the onset of T cell activation/proliferation to the A/JAPAN virus infection preceding the A/PR8 response by 12-24 h. This difference in T cell activation/proliferation correlated with the tempo of accelerated respiratory DC (RDC) migration from the infected lungs to the DLN in response to influenza virus infection, with the migrant RDC responding to the A/JAPAN infection exhibiting a more rapid accumulation in the lymph nodes (i.e., peak migration for A/JAPAN at 18 h, A/PR8 at 24-36 h). Furthermore, in vivo administration of blocking anti-CD62L Ab at various time points before/after infection revealed that the virus-specific CD8+ T cells entered the DLN and activated in a sequential "conveyor belt"-like fashion. These results indicate that the tempo of CD8+ T cell activation/proliferation after viral infection is dependent on the tempo of RDC migration to the DLN and that T cell activation occurs in an ordered sequential fashion.
Collapse
Affiliation(s)
- Heesik Yoon
- Carter Immunology Center, University of Virginia Health Sciences Center, Charlottesville, VA 22908, USA
| | | | | | | |
Collapse
|
139
|
Guarda G, Hons M, Soriano SF, Huang AY, Polley R, Martín-Fontecha A, Stein JV, Germain RN, Lanzavecchia A, Sallusto F. L-selectin-negative CCR7- effector and memory CD8+ T cells enter reactive lymph nodes and kill dendritic cells. Nat Immunol 2007; 8:743-52. [PMID: 17529983 DOI: 10.1038/ni1469] [Citation(s) in RCA: 171] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2007] [Accepted: 04/17/2007] [Indexed: 01/21/2023]
Abstract
T lymphocytes lacking the lymph node-homing receptors L-selectin and CCR7 do not migrate to lymph nodes in the steady state. Instead, we found here that lymph nodes draining sites of mature dendritic cells or adjuvant inoculation recruited L-selectin-negative CCR7- effector and memory CD8+ T cells. This recruitment required CXCR3 expression on T cells and occurred through high endothelial venules in concert with lumenal expression of the CXCR3 ligand CXCL9. In reactive lymph nodes, recruited T cells established stable interactions with and killed antigen-bearing dendritic cells, limiting the ability of these dendritic cells to activate naive CD4+ and CD8+ T cells. The inducible recruitment of blood-borne effector and memory T cells to lymph nodes may represent a mechanism for terminating primary and limiting secondary immune responses.
Collapse
Affiliation(s)
- Greta Guarda
- Institute for Research in Biomedicine, CH-6500 Bellinzona, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Anis MM, Fulton SA, Reba SM, Harding CV, Boom WH. Modulation of naive CD4+ T-cell responses to an airway antigen during pulmonary mycobacterial infection. Infect Immun 2007; 75:2260-8. [PMID: 17296758 PMCID: PMC1865791 DOI: 10.1128/iai.01709-06] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
During pulmonary mycobacterial infection, there is increased trafficking of dendritic cells from the lungs to the draining lymph nodes. We hypothesized that ongoing mycobacterial infection would modulate recruitment and activation of antigen-specific naive CD4+ T cells after airway antigen challenge. BALB/c mice were infected by aerosol with Mycobacterium bovis BCG. At peak bacterial burden in the lungs (4 to 6 weeks postinfection), carboxy-fluorescein diacetate succinimidyl ester-labeled naive ovalbumin-specific DO11.10 T cells were adoptively transferred into infected and uninfected mice. Recipient mice were challenged intranasally with soluble ovalbumin (OVA), and OVA-specific T-cell responses were measured in the lungs, draining mediastinal lymph nodes (MLN), and spleens. OVA challenge resulted in increased activation and proliferation of OVA-specific T cells in the draining MLN of both infected and uninfected mice. However, only BCG-infected mice had prominent OVA-specific T-cell activation, proliferation, and Th1 differentiation in the lungs. BCG infection caused greater distribution of airway OVA to pulmonary dendritic cells and enhanced presentation of OVA peptide by lung CD11c+ cells. Together, these data suggest that an existing pulmonary mycobacterial infection alters the phenotype of lung dendritic cells so that they can activate antigen-specific naive CD4+ T cells in the lungs in response to airway antigen challenge.
Collapse
Affiliation(s)
- Mursalin M Anis
- Department of Pathology, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106-4984, USA
| | | | | | | | | |
Collapse
|
141
|
Sheasley-O’Neill SL, Brinkman CC, Ferguson AR, Dispenza MC, Engelhard VH. Dendritic cell immunization route determines integrin expression and lymphoid and nonlymphoid tissue distribution of CD8 T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 178:1512-22. [PMID: 17237400 PMCID: PMC6046213 DOI: 10.4049/jimmunol.178.3.1512] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Exogenous dendritic cells (bone marrow-derived dendritic cell (BMDC)) display restricted trafficking in vivo after injection into mice, but the route(s) by which they generate gut-homing effector cells is unclear. Mesenteric lymph nodes (LN) and spleen were differentially targeted by i.p. and i.v. administration of BMDC, respectively, whereas mediastinal LN were targeted by both routes. BMDC injected by either route activated CD8(+) T cells to up-regulate both alpha(4)beta(1) and alpha(4)beta(7) integrins. However, the lymphoid compartment in which activation occurred determined their expression kinetics, magnitude, and population distribution. Only T cells activated in mesenteric LN after i.p. immunization expressed high levels of alpha(4)beta(7), which also correlated with localization to small intestine. These alpha(4)beta(7)(high) cells also redistributed to mediastinal LN in a manner sensitive to treatment with alpha(4)beta(7) blocking Abs, but not to mucosal addressin cell adhesion molecule-1 blocking Abs. Our results demonstrate the importance of lymphoid compartment, as dictated by immunization route, in determining integrin expression on activated T cells and their distribution in lymphoid and nonlymphoid tissues.
Collapse
Affiliation(s)
- Stacey L. Sheasley-O’Neill
- Department of Microbiology and Carter Immunology Center, University of Virginia, Charlottesville, VA 22908
| | - C. Colin Brinkman
- Department of Microbiology and Carter Immunology Center, University of Virginia, Charlottesville, VA 22908
| | - Andrew R. Ferguson
- Department of Microbiology and Carter Immunology Center, University of Virginia, Charlottesville, VA 22908
| | | | - Victor H. Engelhard
- Department of Microbiology and Carter Immunology Center, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
142
|
Wikstrom ME, Stumbles PA. Mouse respiratory tract dendritic cell subsets and the immunological fate of inhaled antigens. Immunol Cell Biol 2007; 85:182-8. [PMID: 17262055 DOI: 10.1038/sj.icb.7100039] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
It is widely accepted that tissue dendritic cells (DC) function as immune sentinels by alerting T cells to foreign antigen after delivering and presenting it in the draining lymph nodes. Over the last two decades, studies in animal models, particularly rodents, have demonstrated that respiratory tract DC are crucial for the adaptive immune response to inhaled antigen. Indeed, the fate of inhaled antigen is inextricably linked to the function of respiratory tract DC. In this review, we will discuss the characteristics of respiratory tract DC from mice and recent data that may help to explain their role in the fate of inhaled antigen.
Collapse
Affiliation(s)
- Matthew E Wikstrom
- Telethon Institute for Child Health Research, Centre for Child Health Research, School of Paediatrics and Child Health, University of Western Australia, Perth, Western Australia, Australia.
| | | |
Collapse
|
143
|
Dobrzanski MJ, Reome JB, Hylind JC, Rewers-Felkins KA. CD8-mediated type 1 antitumor responses selectively modulate endogenous differentiated and nondifferentiated T cell localization, activation, and function in progressive breast cancer. THE JOURNAL OF IMMUNOLOGY 2007; 177:8191-201. [PMID: 17114496 DOI: 10.4049/jimmunol.177.11.8191] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD8 T cell-mediated immune responses fall into two distinct types based on effector cell-derived cytokine production. Type I CD8 T cells (Tc1) produce IFN-gamma, whereas type 2 cells (Tc2) secrete IL-4, IL-5, IL-10, and GM-CSF. Using a murine TCR transgenic T cell/breast tumor model, we show that adoptively transferred Ag-specific Tc1 cells are more effective in delaying mammary tumor growth and progression than that of functionally distinct Tc2 cells. Donor Tc1 cells administered 7 days posttumor challenge localized and persisted at sites of primary tumor growth with antitumor responses that were dependent, in part, on effector cell-derived IFN-gamma. Tc1-mediated responses markedly enhanced the appearance and local accumulation of highly differentiated (CD44(high)) CD4 and CD8 endogenous tumor-infiltrating T cells when compared with that of untreated tumor-bearing mice. Conversely, Tc1 cell transfer markedly delayed the appearance of corresponding nondifferentiated (CD44(low)) endogenous T cells. Such cells were acutely activated as defined by coexpression of surface markers associated with TCR engagement (CD69) and early T cell activation (CD25). Moreover, cellular response kinetics appeared to further correlate with the up-regulation of endogenous T cells producing the chemokine IFN-gamma-inducible protein-10 in vivo. This suggested that CD8-mediated type 1 antitumor responses cannot only promote accumulation of distinct endogenous CD4 and CD8 T cell subpopulations, but also facilitate and preferentially modulate their localization kinetics, persistence, states of activation/differentiation, and function within the primary tumor environment at various stages of tumor progression. These studies offer insight into potential mechanisms for enhancing T cell-based immunotherapy in breast cancer.
Collapse
Affiliation(s)
- Mark J Dobrzanski
- Department of Internal Medicine, Texas Tech University School of Medicine, 1400 Wallace Boulevard, Amarillo, TX 79106, USA.
| | | | | | | |
Collapse
|
144
|
Wikstrom ME, Batanero E, Smith M, Thomas JA, von Garnier C, Holt PG, Stumbles PA. Influence of mucosal adjuvants on antigen passage and CD4+ T cell activation during the primary response to airborne allergen. THE JOURNAL OF IMMUNOLOGY 2006; 177:913-24. [PMID: 16818746 DOI: 10.4049/jimmunol.177.2.913] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Ag delivery via the nasal route typically induces tolerance or fails to polarize CD4+ T cell responses unless an adjuvant is provided. To better understand this process, we assessed the effects of two mucosal adjuvants, Escherichia coli LPS and cholera toxin (CT), on Ag passage and T cell activation in the draining lymph nodes (DLN) of BALB/c mice following per nasal administration of the model protein allergen, OVA. We found a range of cell types acquired small amounts of fluorescent OVA in the DLN 4 h after per nasal administration. However, this early uptake was eclipsed by a wave of OVA+CD8alpha(low) dendritic cells that accumulated in the DLN over the next 20 h to become the dominant OVA-processing and -presenting population. Both LPS and CT stimulated increases in CD80 and CD86 expression on OVA+CD8alpha(low) DC. LPS also increased the number of OVA+CD8alpha(low) dendritic cells accumulating in the DLN. When the primary T cell response was examined after adoptive transfer of CD4+ T cells from DO11.10 mice, CT and LPS stimulated surprisingly similar effects on T cell activation and proliferation, IL-4 and IFN-gamma priming, and memory T cell production. Despite these similarities, T cell recipients immunized with CT, but not LPS, developed lung eosinophilia upon secondary OVA challenge. Thus, we found no bias within the DLN in Ag handling or the primary T cell response associated with the eventual Th2 polarization induced by CT, and suggest that additional tissue-specific factors influence the development of allergic disease in the airways.
Collapse
Affiliation(s)
- Matthew E Wikstrom
- Division of Cell Biology, Telethon Institute for Child Health Research, West Perth, WA 6872, Australia.
| | | | | | | | | | | | | |
Collapse
|
145
|
Lefrançois L, Puddington L. Intestinal and pulmonary mucosal T cells: local heroes fight to maintain the status quo. Annu Rev Immunol 2006; 24:681-704. [PMID: 16551263 DOI: 10.1146/annurev.immunol.24.021605.090650] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mucosal immunity in the lung and intestine is controlled by complex multifaceted systems. While mucosal T cells are essential for protection against invading pathogens owing to their proximity to the outside world, powerful systems must also be in place to harness ongoing inflammatory processes. In each site, distinct anatomical structures play key roles in mounting and executing both protective and deleterious mucosal T cell responses. Although analogies can be drawn regarding the immune systems of these two organs, there are substantial dissimilarities necessitated by unique physiologic constraints. Here, we discuss how T cell activation and effector function are generated in the mucosae.
Collapse
Affiliation(s)
- Leo Lefrançois
- Center for Integrative Immunology and Vaccine Research, Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut 06030-1319, USA.
| | | |
Collapse
|
146
|
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
|
147
|
|
148
|
Legge KL, Braciale TJ. Lymph node dendritic cells control CD8+ T cell responses through regulated FasL expression. Immunity 2006; 23:649-59. [PMID: 16356862 DOI: 10.1016/j.immuni.2005.11.006] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2005] [Revised: 09/14/2005] [Accepted: 11/16/2005] [Indexed: 11/15/2022]
Abstract
The lethal outcome of high-dose pulmonary virus infection is thought to reflect high-level, sustained virus replication and associated lung inflammation prior to development of an adaptive immune response. Herein, we demonstrate that the outcome of lethal/sublethal influenza infection instead correlates with the initial virus replication tempo. Furthermore, the magnitude of early lung antiviral CD8+ T cell responses varies inversely with inoculum dose and is controlled by lymph-node-resident dendritic cells (LNDC) through IL-12p40-regulated FasL-dependent T cell apoptosis. These results suggest that the inoculum dose and replication rate of a pathogen entering the respiratory tract may regulate the strength of the adaptive immune response, and the subsequent outcome of infection and that LNDC may serve as regulators (gatekeepers) in the development of CD8+ T cell responses.
Collapse
Affiliation(s)
- Kevin L Legge
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | | |
Collapse
|
149
|
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: 66] [Impact Index Per Article: 3.5] [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
|
150
|
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
|