51
|
Gray PM, Arimilli S, Palmer EM, Parks GD, Alexander-Miller MA. Altered function in CD8+ T cells following paramyxovirus infection of the respiratory tract. J Virol 2005; 79:3339-49. [PMID: 15731228 PMCID: PMC1075682 DOI: 10.1128/jvi.79.6.3339-3349.2005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
For many respiratory pathogens, CD8+ T cells have been shown to play a critical role in clearance. However, there are still many unanswered questions with regard to the factors that promote the most efficacious immune response and the potential for immunoregulation of effector cells at the local site of infection. We have used infection of the respiratory tract with the model paramyxovirus simian virus 5 (SV5) to study CD8+ T-cell responses in the lung. For the present study, we report that over time a population of nonresponsive, virus-specific CD8+ T cells emerged in the lung, culminating in a lack of function in approximately 85% of cells specific for the immunodominant epitope from the viral matrix (M) protein by day 40 postinfection. Concurrent with the induction of nonresponsiveness, virus-specific cells that retained function at later times postinfection exhibited an increased requirement for CD8 engagement. This change was coupled with a nearly complete loss of functional phosphoprotein-specific cells, a response previously shown to be almost exclusively CD8 independent. These studies add to the growing evidence for immune dysregulation following viral infection of the respiratory tract.
Collapse
Affiliation(s)
- Peter M Gray
- Department of Microbiology & Immunology, Room 5108, Gray Building, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | | | | | | | | |
Collapse
|
52
|
Winau F, Sponaas AM, Weber S, Schwierzeck V, Winter R, Hurwitz R, Kaufmann SHE. Scant activation of CD8 T?cells by antigen loaded on heat shock protein. Eur J Immunol 2005; 35:1046-55. [PMID: 15739167 DOI: 10.1002/eji.200425603] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Heat shock proteins (HSP) not only function as chaperones for denatured proteins but also for antigenic peptides, thus inducing protective T cell responses. Here we show that vaccination with peptide-loaded HSP70 causes initial interferon-gamma production by murine CD8 T cells but no T cell expansion. These CD8 T cells lacked cytotoxic activity in vitro and in vivo, which was not due to apoptosis. Restimulation with peptide-pulsed dendritic cells both bypassed the proliferative block and suspended the non-protective state of CD8 T lymphocytes in an infection model with the bacterial pathogen, Listeria monocytogenes. Cotransfer of antigen-specific CD4 T cells circumvented the proliferative arrest of CD8 T cells. Our data suggest that HSP vaccines induce CD8 T cell unresponsiveness unless proficient help is provided. Assuming that this model reflects the antigenically experienced human condition where immunological space is restricted and any T cell response possibly leads to suppression of heterologous reactions, our findings bear implications for rational vaccination protocols including those for immunocompromised patients.
Collapse
Affiliation(s)
- Florian Winau
- Department of Immunology, Max-Planck-Institute for Infection Biology, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
53
|
Redmond WL, Marincek BC, Sherman LA. Distinct requirements for deletion versus anergy during CD8 T cell peripheral tolerance in vivo. THE JOURNAL OF IMMUNOLOGY 2005; 174:2046-53. [PMID: 15699134 DOI: 10.4049/jimmunol.174.4.2046] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Activation of naive T cells by quiescent APCs results in tolerance through deletion and anergy. The underlying basis for these distinct fates is unclear. Using clone 4 TCR transgenic animals as a source of naive CD8 T cells, we examined the requirements for peripheral deletion in vivo. Our results demonstrate that independent of the amount of Ag used for stimulation, a single dose was insufficient to achieve complete clonal deletion. Instead, further antigenic exposure was required to completely eliminate all of the activated T cells. Additionally, consecutive stimulations with low doses of Ag were highly effective in promoting deletion. In contrast, although stimulation with high doses of Ag initially led to the apoptosis of many of the activated T cells, it induced hyporesponsiveness in a portion of the responding cells, thereby sparing them from further activation and deletion. These data explain why some conditions promote tolerance through clonal deletion whereas others promote anergy. Furthermore, these data provide a framework to devise protocols for effective deletion of potentially autoreactive T cells.
Collapse
Affiliation(s)
- William L Redmond
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
54
|
Valenzuela JO, Hammerbeck CD, Mescher MF. Cutting edge: Bcl-3 up-regulation by signal 3 cytokine (IL-12) prolongs survival of antigen-activated CD8 T cells. THE JOURNAL OF IMMUNOLOGY 2005; 174:600-4. [PMID: 15634875 DOI: 10.4049/jimmunol.174.2.600] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Clonal expansion of T cells requires cell division and survival during the proliferative phase of the response. Naive murine CD8 T cells responding to Ag and costimulation undergo an abortive response characterized by impaired clonal expansion, failure to develop effector functions, and long-term tolerance. A third signal provided by IL-12 is required for full expansion, activation, and establishment of memory. The enhanced survival, and thus clonal expansion, supported by IL-12 is not due to increased Bcl-2 or Bcl-x(L) expression; both are maximally activated by signals 1 and 2. In contrast, Bcl-3, recently shown to enhance survival when ectopically expressed in T cells, is increased only when IL-12 is present. Furthermore, examination of Bcl-3-deficient CD8 T cells demonstrates that the increased survival caused by IL-12 depends upon Bcl-3. The time courses of expression suggest that Bcl-2 and Bcl-x(L) promote survival early in the response, whereas Bcl-3 acts later in the response.
Collapse
Affiliation(s)
- Javier O Valenzuela
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
55
|
Dudda JC, Lembo A, Bachtanian E, Huehn J, Siewert C, Hamann A, Kremmer E, Förster R, Martin SF. Dendritic cells govern induction and reprogramming of polarized tissue-selective homing receptor patterns of T cells: important roles for soluble factors and tissue microenvironments. Eur J Immunol 2005; 35:1056-65. [PMID: 15739162 DOI: 10.1002/eji.200425817] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Tissue-selective homing is established during naive T cell activation by the tissue microenvironment and tissue-specific dendritic cells (DC). The factors driving induction and maintenance of T cell homing patterns are still largely unknown. Here we show that soluble factors produced during the interaction of T cells with CD11c(+) DC isolated from skin- or small intestine-associated tissues differentially modulate expression of the corresponding tissue-selective homing receptors (E-selectin ligands and alpha4beta7 integrin/CCR9, respectively) on murine CD8(+) T cells. Injection of tissue-specific DC via different routes induces T cells with homing receptors characteristic of the corresponding local tissue microenvironment, independent of the origin of the DC. These data indicate an important role for signals delivered in trans. Moreover, DC can reprogram the homing receptor expression on T cells previously polarized in vitro for homing to skin or small intestine. Importantly, skin-homing memory T cells stimulated directly ex vivo can also be reprogrammed by intestinal DC to a gut-homing phenotype. Our results show that tissue-selective homing receptor expression on effector and memory T cells is governed by inductive as well as suppressive signals from both DC and tissue microenvironments.
Collapse
Affiliation(s)
- Jan C Dudda
- Clinical Research Group Allergology, Department of Dermatology, University of Freiburg, Freiburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
56
|
Redmond WL, Sherman LA. Peripheral Tolerance of CD8 T Lymphocytes. Immunity 2005; 22:275-84. [PMID: 15780985 DOI: 10.1016/j.immuni.2005.01.010] [Citation(s) in RCA: 177] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2004] [Revised: 01/10/2005] [Accepted: 01/13/2005] [Indexed: 01/18/2023]
Abstract
Whereas high-avidity recognition of peptide-MHC complexes by developing T cells in the thymus results in deletion and promotes self-tolerance, such recognition by mature T cells in the periphery results in activation and clonal expansion. This dichotomy represents the basis of a dilemma that has stumped immunologists for many years, how are self-specific T cells tolerized in the periphery? There appear to be two important criteria used to achieve this goal. The first is that in the absence of inflammatory pathogens, tolerance is promoted when T cells recognize antigen presented by quiescent dendritic cells (DCs) expressing low levels of costimulatory molecules. A second critical factor that defines "self" and drives tolerance through deletion, anergy, or suppression is the persistence of antigen.
Collapse
Affiliation(s)
- William L Redmond
- Department of Immunology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | |
Collapse
|
57
|
Guichard G. MHC ligands as potential therapeutics in the treatment of autoimmune and allergic diseases. Expert Opin Ther Pat 2005. [DOI: 10.1517/13543776.7.1.29] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
58
|
Engelhardt KR, Richter K, Baur K, Staeheli P, Hausmann J. The functional avidity of virus-specific CD8+ T?cells is down-modulated in Borna disease virus-induced immunopathology of the central nervous system. Eur J Immunol 2005; 35:487-97. [PMID: 15627979 DOI: 10.1002/eji.200425232] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Borna disease virus (BDV) infection of the central nervous system (CNS) leads to severe neurological symptoms in susceptible MRL mice. The disease is mainly mediated by CD8+ T cells specific for the immunodominant epitope TELEISSI in the BDV nucleoprotein. In this study, TELEISSI/MHC class I tetramers were used to directly visualize antigen-specific CD8+ T cells. We found that on average approximately 30% of the ex vivo analyzed CD8+ T cells in the CNS of diseased mice were specific for TELEISSI. Unexpectedly, the frequency of tetramer-reactive brain-derived CD8+ T cells doubled following overnight culture in the absence of antigen. The majority of CD8+ T cells showed enhanced tetramer binding without up-regulation of T cell receptor surface expression. The frequency of IFN-gamma-secreting CD8+ T cells after antigen-specific stimulation was higher in overnight cultures than in freshly isolated BDV-specific brain lymphocytes, and enhanced tetramer binding correlated with elevated sensitivity to lower levels of peptide antigen in cytotoxicity assays. These results indicate that the functional avidity of virus-specific CD8+ T cells was down-modulated in vivo. Thus, quantification of tissue-infiltrating CD8+ T cells by the tetramer technique must be interpreted with caution as it may underestimate the real frequency of antigen-specific CD8+ T cells.
Collapse
Affiliation(s)
- Karin R Engelhardt
- Department of Virology, Institute of Medical Microbiology and Hygiene, University of Freiburg, Freiburg, Germany
| | | | | | | | | |
Collapse
|
59
|
Kissenpfennig A, Aït-Yahia S, Clair-Moninot V, Stössel H, Badell E, Bordat Y, Pooley JL, Lang T, Prina E, Coste I, Gresser O, Renno T, Winter N, Milon G, Shortman K, Romani N, Lebecque S, Malissen B, Saeland S, Douillard P. Disruption of the langerin/CD207 gene abolishes Birbeck granules without a marked loss of Langerhans cell function. Mol Cell Biol 2005; 25:88-99. [PMID: 15601833 PMCID: PMC538791 DOI: 10.1128/mcb.25.1.88-99.2005] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2004] [Revised: 09/03/2004] [Accepted: 10/10/2004] [Indexed: 11/20/2022] Open
Abstract
Langerin is a C-type lectin expressed by a subset of dendritic leukocytes, the Langerhans cells (LC). Langerin is a cell surface receptor that induces the formation of an LC-specific organelle, the Birbeck granule (BG). We generated a langerin(-/-) mouse on a C57BL/6 background which did not display any macroscopic aberrant development. In the absence of langerin, LC were detected in normal numbers in the epidermis but the cells lacked BG. LC of langerin(-/-) mice did not present other phenotypic alterations compared to wild-type littermates. Functionally, the langerin(-/-) LC were able to capture antigen, to migrate towards skin draining lymph nodes, and to undergo phenotypic maturation. In addition, langerin(-/-) mice were not impaired in their capacity to process native OVA protein for I-A(b)-restricted presentation to CD4(+) T lymphocytes or for H-2K(b)-restricted cross-presentation to CD8(+) T lymphocytes. langerin(-/-) mice inoculated with mannosylated or skin-tropic microorganisms did not display an altered pathogen susceptibility. Finally, chemical mutagenesis resulted in a similar rate of skin tumor development in langerin(-/-) and wild-type mice. Overall, our data indicate that langerin and BG are dispensable for a number of LC functions. The langerin(-/-) C57BL/6 mouse should be a valuable model for further functional exploration of langerin and the role of BG.
Collapse
MESH Headings
- 9,10-Dimethyl-1,2-benzanthracene
- Animals
- Antigens/metabolism
- Antigens, Surface/genetics
- Antigens, Surface/physiology
- Blastocyst/metabolism
- CD4-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/metabolism
- Carcinogens
- Cell Movement
- Cell Physiological Phenomena
- Cytoplasmic Granules/metabolism
- Dendritic Cells
- Dose-Response Relationship, Drug
- Electroporation
- Embryo, Mammalian/cytology
- Flow Cytometry
- Genetic Vectors
- Immunohistochemistry
- Islets of Langerhans/cytology
- Islets of Langerhans/physiology
- Kinetics
- Langerhans Cells/cytology
- Lectins/metabolism
- Lectins, C-Type/genetics
- Lectins, C-Type/physiology
- Lymph Nodes/metabolism
- Mannose-Binding Lectins/genetics
- Mannose-Binding Lectins/physiology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Transgenic
- Microscopy, Electron
- Models, Genetic
- Mutagenesis
- Mutation
- Neoplasms/chemically induced
- Ovalbumin/metabolism
- Phenotype
- Stem Cells/cytology
Collapse
Affiliation(s)
- Adrien Kissenpfennig
- Centre d'Immunologie de Marseille-Luminy, INSERM-CNRS-Université de la Méditerranee, Parc Scientifique de Luminy, Marseille, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Piaggio E, Hartemann-Heurtier A, Cabarrocas J, Desbois S, Mars LT, Zappulla JP, Liblau RS. Maintaining or breaking CD8+ T-cell tolerance to beta islet cell antigens: lessons from transgenic mouse models. J Autoimmun 2004; 22:115-20. [PMID: 14987739 DOI: 10.1016/j.jaut.2003.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Eliane Piaggio
- INSERM U563, Purpan University Hospital, Place Dr Baylac, Toulouse 31000, France
| | | | | | | | | | | | | |
Collapse
|
61
|
Rowley TF, Al-Shamkhani A. Stimulation by soluble CD70 promotes strong primary and secondary CD8+ cytotoxic T cell responses in vivo. THE JOURNAL OF IMMUNOLOGY 2004; 172:6039-46. [PMID: 15128787 DOI: 10.4049/jimmunol.172.10.6039] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Identification of the signals required for optimal differentiation of naive CD8(+) T cells into effector and memory cells is critical for the design of effective vaccines. In this study we demonstrate that CD27 stimulation by soluble CD70 considerably enhances the magnitude and quality of the CD8(+) T cell response. Stimulation with soluble CD70 in the presence of Ag significantly enhanced the proliferation of CD8(+) T cells and their ability to produce IL-2 and IFN-gamma in vitro. Administration of Ag and soluble CD70 resulted in a massive (>300-fold) expansion of Ag-specific CD8(+) T cells in vivo, which was due to the enhanced proliferation and survival of activated T cells. In mice that received Ag and soluble CD70, CD8(+) T cells developed into effectors with direct ex vivo cytotoxicity. Furthermore, unlike peptide immunization, which resulted in a diminished response after rechallenge, CD27 stimulation during the primary challenge evoked a strong secondary response upon rechallenge with the antigenic peptide. Thus, in addition to increasing the frequency of primed Ag-specific T cells, CD27 signaling during the primary response instills a program of differentiation that allows CD8(+) T cells to overcome a state of unresponsiveness. Taken together these results demonstrate that soluble CD70 has potent in vivo adjuvant effects for CD8(+) T cell responses.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/metabolism
- Adjuvants, Immunologic/physiology
- Adoptive Transfer
- Animals
- Antigens, CD/administration & dosage
- Antigens, CD/metabolism
- Antigens, CD/physiology
- CD27 Ligand
- Cell Differentiation/immunology
- Cell Line
- Cytotoxicity, Immunologic
- Epitopes, T-Lymphocyte/administration & dosage
- Epitopes, T-Lymphocyte/immunology
- Humans
- Immunization, Secondary
- Lymphocyte Activation
- Membrane Proteins/administration & dosage
- Membrane Proteins/metabolism
- Membrane Proteins/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Ovalbumin/administration & dosage
- Ovalbumin/immunology
- Peptide Fragments/administration & dosage
- Peptide Fragments/immunology
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/metabolism
- Recombinant Fusion Proteins/physiology
- Signal Transduction/immunology
- Solubility
- T-Lymphocytes, Cytotoxic/cytology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/transplantation
- Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
- Tumor Necrosis Factor Receptor Superfamily, Member 7/physiology
Collapse
Affiliation(s)
- Tania F Rowley
- Tenovus Research Laboratory, Cancer Sciences Division, School of Medicine, Southampton General Hospital, Southampton, United Kingdom
| | | |
Collapse
|
62
|
Jeon MS, Atfield A, Venuprasad K, Krawczyk C, Sarao R, Elly C, Yang C, Arya S, Bachmaier K, Su L, Bouchard D, Jones R, Gronski M, Ohashi P, Wada T, Bloom D, Fathman CG, Liu YC, Penninger JM. Essential Role of the E3 Ubiquitin Ligase Cbl-b in T Cell Anergy Induction. Immunity 2004; 21:167-77. [PMID: 15308098 DOI: 10.1016/j.immuni.2004.07.013] [Citation(s) in RCA: 276] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2003] [Revised: 06/01/2004] [Accepted: 06/07/2004] [Indexed: 11/25/2022]
Abstract
Antigen-specific immunotolerance limits the expansion of self-reactive T cells involved in autoimmune diseases. Here, we show that the E3 ubiquitin ligase Cbl-b is upregulated in T cells after tolerizing signals. Loss of Cbl-b in mice results in impaired induction of T cell tolerance both in vitro and in vivo. Importantly, rechallenge of Cbl-b mutant mice with the tolerizing antigen results in massive lethality. Moreover, ablation of Cbl-b resulted in exacerbated autoimmunity. Mechanistically, loss of Cbl-b rescues reduced calcium mobilization of anergic T cells, which was attributed to Cbl-b-mediated regulation of PLCgamma-1 phosphorylation. Our results show a critical role for Cbl-b in the regulation of peripheral tolerance and anergy of T cells.
Collapse
Affiliation(s)
- Myung-Shin Jeon
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, San Diego, California 92121, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Koschella M, Voehringer D, Pircher H. CD40 ligation in vivo induces bystander proliferation of memory phenotype CD8 T cells. THE JOURNAL OF IMMUNOLOGY 2004; 172:4804-11. [PMID: 15067057 DOI: 10.4049/jimmunol.172.8.4804] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Injection of agonistic anti-CD40 Abs into mice has been shown to amplify weak CD8 T cell responses to poorly immunogenic compounds and to convert T cell tolerance to T cell priming. In this study we demonstrate that anti-CD40 treatment of C57BL/6 mice, without Ag delivery, led to a marked increase in the number of memory phenotype CD4 and CD8 T cells. Adoptive transfer experiments using CD40-deficient hosts further revealed that the proliferative response of memory T cells, induced by systemic CD40 signaling, was dependent on CD40 expression of host APCs. CD40 ligation in vivo induced vigorous cell division of both memory phenotype and bona fide virus-specific memory CD8 T cells in a partially IL-15-dependent manner. However, only memory phenotype, but not Ag-experienced memory CD8 T cells increased in cell number after anti-CD40 treatment in vivo. Taken together our data show that activation of APC via CD40 induces a marked bystander proliferation of memory phenotype T cells. In addition, we demonstrate that bona fide Ag-experienced memory CD8 T cells respond differently to anti-CD40-induced signals than memory phenotype CD8 T cells.
Collapse
Affiliation(s)
- Marie Koschella
- Institute of Medical Microbiology and Hygiene, Department of Immunology, University of Freiburg, Freiburg, Germany
| | | | | |
Collapse
|
64
|
Dudda JC, Denfeld RW, Simon JC, Martin SF. UVB-irradiated dendritic cells fail to tolerize murine CD8 naïve or effector T cells. J Invest Dermatol 2004; 122:945-52. [PMID: 15102085 DOI: 10.1111/j.0022-202x.2004.22423.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
UVB radiation has been shown to induce T cell tolerance most likely via modulation of the function of antigen-presenting cells like dendritic cells (DC), which are therefore of interest for vaccination therapy. Since little is known about the effects of UVB-irradiated dendritic cells (UVB-DC) on CD8(+) T cells, which are the dominant effectors in various allergic and autoimmune diseases, we have investigated the potential of low dose UVB (100-200 J per m(2)) irradiated bone marrow-derived dendritic cells to induce tolerance in murine CD8(+) T cells specific for the contact allergen trinitrophenyl (TNP) or for a viral peptide. In contrast to the previously reported successful tolerization of primed CD4(+) Th1 cells, neither naïve CD8(+) T cells nor CD8(+) Tc1 effector cells or established CD8(+) T cell clones could be tolerized by TNP-modified or peptide-pulsed UVB-DC in vitro or in vivo. We observed, however, a reduced capacity of UVB-DC to prime naïve CD8(+) T cells. Our data demonstrate an important difference in the susceptibility of CD4(+) and CD8(+) T cells for tolerance induction using low-dose UVB-irradiated DC and have implications for DC therapy of CD8(+) T cell-mediated diseases.
Collapse
Affiliation(s)
- Jan C Dudda
- Clinical Research Group Allergology, Department of Dermatology, University of Freiburg, Freiburg, Germany
| | | | | | | |
Collapse
|
65
|
Roth E, Pircher H. IFN-gamma promotes Fas ligand- and perforin-mediated liver cell destruction by cytotoxic CD8 T cells. THE JOURNAL OF IMMUNOLOGY 2004; 172:1588-94. [PMID: 14734739 DOI: 10.4049/jimmunol.172.3.1588] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
To study liver cell damage by CTL, CD8 T cells from P14 TCR transgenic (tg) mice specific for the gp33 epitope of lymphocytic choriomeningitis virus with either deficiency in IFN-gamma (P14.IFN-gamma(null)), functional Fas ligand (P14.gld), or perforin (P14.PKO) were transferred into H8 tg mice ubiquitously expressing gp33 Ag. Treatment of H8 recipient mice with agonistic anti-CD40 Abs induced vigorous expansion of the transferred P14 T cells and led to liver cell destruction determined by increase of glutamate dehydrogenase serum levels and induction of caspase-3 in hepatocytes. Liver injury was mediated by the Fas/Fas ligand (FasL) pathway and by perforin, because P14.gld and P14.PKO T cells failed to induce increased glutamate dehydrogenase levels despite strong in vivo proliferation. In addition, H8 tg mice lacking Fas were resistant to the pathogenic effect of P14 T cells. Besides FasL and perforin, IFN-gamma was also required for liver cell damage, because P14.IFN-gamma(null) T cells adoptively transferred into H8 mice failed to induce disease. Moreover, Fas expression on hepatocytes from H8 recipient mice was increased after transfer of wild-type compared with P14.IFN-gamma(null) T cells, and wild-type P14 T cells expressed higher levels of FasL than P14 T cells lacking IFN-gamma. Thus, our data suggest that IFN-gamma released by activated CD8 T cells upon Ag contact facilitates liver cell destruction.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Cytotoxicity, Immunologic/genetics
- Fas Ligand Protein
- Female
- Hepatitis, Animal/genetics
- Hepatitis, Animal/immunology
- Hepatitis, Animal/pathology
- Hepatocytes/immunology
- Hepatocytes/metabolism
- Interferon-gamma/deficiency
- Interferon-gamma/genetics
- Interferon-gamma/metabolism
- Interferon-gamma/physiology
- Ligands
- Lymphocyte Activation/genetics
- Male
- Membrane Glycoproteins/biosynthesis
- Membrane Glycoproteins/deficiency
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Perforin
- Pore Forming Cytotoxic Proteins
- Spleen/immunology
- Spleen/pathology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Cytotoxic/transplantation
- Up-Regulation/genetics
- Up-Regulation/immunology
- fas Receptor/biosynthesis
- fas Receptor/metabolism
Collapse
Affiliation(s)
- Evelyn Roth
- Institute for Medical Microbiology and Hygiene, Department of Immunology, University of Freiburg, Freiburg, Germany
| | | |
Collapse
|
66
|
Dudda JC, Simon JC, Martin S. Dendritic cell immunization route determines CD8+ T cell trafficking to inflamed skin: role for tissue microenvironment and dendritic cells in establishment of T cell-homing subsets. THE JOURNAL OF IMMUNOLOGY 2004; 172:857-63. [PMID: 14707056 DOI: 10.4049/jimmunol.172.2.857] [Citation(s) in RCA: 152] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The effector/memory T cell pool branches in homing subsets selectively trafficking to organs such as gut or skin. Little is known about the critical factors in the generation of skin-homing CD8+ T cells, although they are crucial effectors in skin-restricted immune responses such as contact hypersensitivity and melanoma defense. In this study, we show that intracutaneous, but not i.v. injection of bone marrow-derived dendritic cells induced skin-homing CD8+ T cells with up-regulated E-selectin ligand expression and effector function in contact hypersensitivity. The skin-homing potential and E-selectin ligand expression remained stable in memory phase without further Ag contact. In contrast, i.p. injection induced T cells expressing the gut-homing integrin alpha(4)beta(7). Although differential expression of these adhesion molecules was strictly associated with the immunization route, the postulated skin-homing marker CCR4 was transiently up-regulated in all conditions. Interestingly, dendritic cells from different tissues effectively induced the corresponding homing markers on T cells in vitro. Our results suggest a crucial role for the tissue microenvironment and dendritic cells in the instruction of T cells for tissue-selective homing and demonstrate that Langerhans cells are specialized to target T cells to inflamed skin.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/pathology
- Cell Movement/immunology
- Chemokines, CC/metabolism
- Dendritic Cells/immunology
- Dendritic Cells/pathology
- Dendritic Cells/transplantation
- Dermatitis, Contact/immunology
- Dermatitis, Contact/pathology
- Immunologic Memory
- Inflammation/immunology
- Injections, Intradermal
- Injections, Intravenous
- Membrane Glycoproteins/biosynthesis
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Organ Specificity/immunology
- Picryl Chloride/administration & dosage
- Picryl Chloride/immunology
- Receptors, CCR4
- Receptors, Chemokine/biosynthesis
- Receptors, Fibroblast Growth Factor/biosynthesis
- Sialoglycoproteins
- Skin/immunology
- Skin/pathology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/pathology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
Collapse
Affiliation(s)
- Jan C Dudda
- Clinical Research Group Allergology, Department of Dermatology, University of Freiburg, Hauptstrasse 7, D-79104 Freiburg, Germany
| | | | | |
Collapse
|
67
|
Redmond WL, Hernandez J, Sherman LA. Deletion of naive CD8 T cells requires persistent antigen and is not programmed by an initial signal from the tolerogenic APC. THE JOURNAL OF IMMUNOLOGY 2004; 171:6349-54. [PMID: 14662832 DOI: 10.4049/jimmunol.171.12.6349] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Activation of naive CD8 T cells in vivo requires the recognition of cognate peptide-MHC complexes on APCs. Depending upon the activation status of the APC, such recognition will promote either a vigorous immune response or T cell tolerance and deletion. Recent studies suggest that the initial signals provided by APCs are sufficient to program the proliferation of naive CD8 T cells and their differentiation into effector cells. In this study, we sought to determine whether an initial encounter with tolerogenic APCs was sufficient to program deletion of naive CD8 T cells. Surprisingly, we find that regardless of whether naive CD8 T cells were stimulated by activated or quiescent APCs, transfer of the activated T cells into an Ag-free host was sufficient to ensure survival. Thus, although the extent of clonal expansion and development of effector function is determined by the activation status of the stimulatory APC, peripheral clonal deletion requires persistent Ag and is not determined by the initial stimulatory event.
Collapse
Affiliation(s)
- William L Redmond
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
68
|
Millar DG, Garza KM, Odermatt B, Elford AR, Ono N, Li Z, Ohashi PS. Hsp70 promotes antigen-presenting cell function and converts T-cell tolerance to autoimmunity in vivo. Nat Med 2003; 9:1469-76. [PMID: 14625545 DOI: 10.1038/nm962] [Citation(s) in RCA: 207] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2003] [Accepted: 10/28/2003] [Indexed: 11/09/2022]
Abstract
Pathogens or pathogen-associated molecular patterns can signal to cells of the innate immune system and trigger effective adaptive immunity. However, relatively little is known about how the innate immune system detects tissue injury or necrosis. Evidence suggests that the release of heat-shock proteins (HSPs) may provide adjuvant-like signals, but the ability of HSPs to promote activation or tolerance in vivo has not been addressed. In this study we show that Hsp70 promotes dendritic cell (DC) function and, together with antigen, triggers autoimmune disease in vivo.
Collapse
Affiliation(s)
- Douglas G Millar
- Departments of Immunology and Medical Biophysics, University Health Network, Ontario Cancer Institute, Toronto, Ontario M5G 2M9, Canada
| | | | | | | | | | | | | |
Collapse
|
69
|
Cytotoxic T lymphocytes generated by short-term in vitro TCR stimulation in the presence of IL-4 are therapeutically effective against B16 melanoma. J Biomed Sci 2003. [DOI: 10.1007/bf02256315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
70
|
Zarei S, Arrighi JF, Ongaro G, Calzascia T, Haller O, Frossard C, Piguet V, Walker PR, Hauser C. Efficient Induction of CD8 T-Associated Immune Protection by Vaccination with mRNA Transfected Dendritic Cells. J Invest Dermatol 2003; 121:745-50. [PMID: 14632191 DOI: 10.1046/j.1523-1747.2003.12492.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Dendritic cells are excellent targets for antigen-specific immune intervention. Here we attempted to introduce a CD8 T cell-dependent epitope into dendritic cells for presentation on major histocompatibility complex class I and induction of immunity. Murine bone-marrow-derived dendritic cells were subjected to electroporation with mRNA transcribed in vitro from a plasmid encoding lymphocytic choriomeningitis virus glycoprotein or enhanced green fluorescent protein under the control of a T7 promotor. The transfection efficiency of dendritic cells was 22 to 40%. Maturation was not inhibited by the electroporation. Dendritic cells electroporated with the appropriate antigen induced cell number-dependent in vitro proliferation in CD8 T cells expressing a transgenic receptor recognizing the 33 to 41 sequence of lymphocytic choriomeningitis virus glycoprotein in association with H-2Kb/Db, indicating correct synthesis, processing, and presentation of the epitope. Naive C57BL/6 mice vaccinated with electroporated dendritic cells and challenged with lymphocytic choriomeningitis virus were protected. Vaccination induced epitope-specific T cells as assessed by tetramer staining in blood and spleen. These results indicate that targeting dendritic cells with antigen-encoding mRNA can induce antigen-specific CD8 T cell responses as well as protective anti-viral immunity in vivo. Targeting dendritic cells with antigen-encoding mRNA may find wider application for immune intervention in disorders such as autoimmunity and cancer.
Collapse
Affiliation(s)
- Shohreh Zarei
- Allergy Unit, Division of Immunology and Allergy, University Hospital, Geneva Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Abstract
Dendritic cells (DCs) have several functions in innate and adaptive immunity. In addition, there is increasing evidence that DCs in situ induce antigen-specific unresponsiveness or tolerance in central lymphoid organs and in the periphery. In the thymus DCs generate tolerance by deleting self-reactive T cells. In peripheral lymphoid organs DCs also induce tolerance to antigens captured by receptors that mediate efficient uptake of proteins and dying cells. Uptake by these receptors leads to the constitutive presentation of antigens on major histocompatibility complex (MHC) class I and II products. In the steady state the targeting of DC antigen capture receptors with low doses of antigens leads to deletion of the corresponding T cells and unresponsiveness to antigenic rechallenge with strong adjuvants. In contrast, if a stimulus for DC maturation is coadministered with the antigen, the mice develop immunity, including interferon-gamma-secreting effector T cells and memory T cells. There is also new evidence that DCs can contribute to the expansion and differentiation of T cells that regulate or suppress other immune T cells. One possibility is that distinct developmental stages and subsets of DCs and T cells can account for the different pathways to peripheral tolerance, such as deletion or suppression. We suggest that several clinical situations, including autoimmunity and certain infectious diseases, can be influenced by the antigen-specific tolerogenic role of DCs.
Collapse
Affiliation(s)
- Ralph M Steinman
- Laboratory of Cellular Physiology and Immunology, The Rockefeller University, New York, New York 10021-6399, USA.
| | | | | |
Collapse
|
72
|
Storni T, Ruedl C, Renner WA, Bachmann MF. Innate immunity together with duration of antigen persistence regulate effector T cell induction. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:795-801. [PMID: 12847247 DOI: 10.4049/jimmunol.171.2.795] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Proliferation of T cells is important for the expansion of specific T cell clones during immune responses. In addition, for the establishment of protective immunity against viruses, bacteria, and tumors, the expanded T cells must differentiate into effector T cells. Here we show that effector T cell generation is driven by activation of APCs and duration of antigenic stimulation. Adoptively transferred TCR-transgenic T cells extensively proliferated upon immunization. However, these T cells failed to differentiate into effector cells and died within 1 wk after immunization unless antigenic peptides persisted for >1 day or were presented by activated APCs. The induction of protective immunity in a nontransgenic system was more stringent, since activation of APCs or prolonged Ag persistence alone was not sufficient to drive immunity. In contrast, Ag had to be presented for several days by activated APCs to trigger protective T cell responses. Thus, activation of APCs and duration of Ag presentation together regulate the induction of protective T cell responses.
Collapse
MESH Headings
- Animals
- Antigen Presentation/genetics
- Antigen Presentation/immunology
- Antigens, CD/biosynthesis
- Antigens, Differentiation, T-Lymphocyte/biosynthesis
- Antigens, Viral/administration & dosage
- Antigens, Viral/immunology
- Antigens, Viral/metabolism
- Biomarkers/analysis
- Cell Division/genetics
- Cell Division/immunology
- Female
- Glycoproteins/administration & dosage
- Glycoproteins/immunology
- Glycoproteins/metabolism
- Hyaluronan Receptors/biosynthesis
- Immunity, Innate/genetics
- Lectins, C-Type
- Lymphocyte Activation/genetics
- Lymphocytic choriomeningitis virus/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Peptide Fragments/administration & dosage
- Peptide Fragments/immunology
- Peptide Fragments/metabolism
- Receptors, Interleukin-2/biosynthesis
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/virology
- Time Factors
- Up-Regulation/genetics
- Up-Regulation/immunology
- Viral Proteins/administration & dosage
- Viral Proteins/immunology
- Viral Proteins/metabolism
- Viral Vaccines/administration & dosage
- Viral Vaccines/immunology
- Viral Vaccines/metabolism
- Virion/immunology
- Virion/metabolism
Collapse
Affiliation(s)
- Tazio Storni
- Cytos Biotechnology, Schlieren-Zurich, Switzerland
| | | | | | | |
Collapse
|
73
|
Curtsinger JM, Lins DC, Mescher MF. Signal 3 determines tolerance versus full activation of naive CD8 T cells: dissociating proliferation and development of effector function. J Exp Med 2003; 197:1141-51. [PMID: 12732656 PMCID: PMC2193970 DOI: 10.1084/jem.20021910] [Citation(s) in RCA: 369] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Activation of naive CD8 T cells to undergo clonal expansion and develop effector function requires three signals: (a) Ag, (b) costimulation, and (c) IL-12 or adjuvant. The requirement for the third signal to stimulate Ag-dependent proliferation is variable, making the greatest contribution when Ag levels are low. At high Ag levels, extensive proliferation can occur in vitro or in vivo in the absence of a third signal. However, despite having undergone the same number of divisions, cells that expand in the absence of a third signal fail to develop cytolytic effector function. Thus, proliferation and development of cytolytic function can be fully uncoupled. Furthermore, these cells are rendered functionally tolerant in vivo, in that subsequent restimulation with a potent stimulus results in limited clonal expansion, impaired IFN-gamma production, and no cytolytic function. Thus, the presence or absence of the third signal appears to be a critical variable in determining whether stimulation by Ag results in tolerance versus development of effector function and establishment of a responsive memory population.
Collapse
Affiliation(s)
- Julie M Curtsinger
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota, MN 55455, USA
| | | | | |
Collapse
|
74
|
Dileo J, Banerjee R, Whitmore M, Nayak JV, Falo LD, Huang L. Lipid-protamine-DNA-mediated antigen delivery to antigen-presenting cells results in enhanced anti-tumor immune responses. Mol Ther 2003; 7:640-8. [PMID: 12718907 DOI: 10.1016/s1525-0016(03)00064-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Vaccination with antigenic peptides encoding tumor antigens has the potential to be an effective treatment for cancer. To induce tumor-specific cellular immune responses, a peptide antigen must be presented by antigen-presenting cells (APCs) to T-cells in the lymphatic tissues. Effective in vivo delivery of peptide antigens to APCs has been problematic. Here we use a model antigen from the HPV16 E7 protein to formulate LPD/E7 particles that upon iv administration are internalized by CD11c(+) and CD11b(+) cells in the marginal zone of the spleen. Either iv or sc vaccination with LPD/E7 particles induces E7-specific CTL responses stronger than those obtained using previously described liposome/peptide strategies and prevents the establishment of E7-expressing tumors. Furthermore, the administration of LPD/E7 particles to tumor-bearing mice caused complete tumor regression in 100% of the treated animals. Based on these studies, the entrapment of peptide antigens inside LPD particles may be an effective and generally applicable strategy for the enhancement of peptide vaccine potency.
Collapse
Affiliation(s)
- John Dileo
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | | | | | |
Collapse
|
75
|
Köller MD, Kiener HP, Aringer M, Graninger WB, Meuer S, Samstag Y, Smolen JS. Functional and molecular aspects of transient T cell unresponsiveness: role of selective interleukin-2 deficiency. Clin Exp Immunol 2003; 132:225-31. [PMID: 12699409 PMCID: PMC1808700 DOI: 10.1046/j.1365-2249.2003.02150.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Defects of T cell (Tc) proliferation have been demonstrated in several autoimmune diseases. Detailed mechanisms governing activation and proliferation of Tc are still not completely known. Here we show that under certain conditions human peripheral blood lymphocytes, once activated by anti-CD3, fail to respond to a subsequent restimulation via the Tc-receptor. Peripheral blood mononuclear cells (PBMC) were preactivated by anti-CD3 for 96 h following restimulation by anti-CD3, interleukin (IL)-2 and other mitogens. In control experiments unstimulated PBMC were incubated in medium alone. Immunophenotypes were analysed by flow cytometry. Cytokine production was determined by reverse transcription-polymerase chain reaction and intracellular signalling protein contents of Tc were compared by Western blotting. Furthermore, apoptosis was detected by terminal deoxyribose transferase-mediated deoxyuridine triphosphate nick end labelling assay. Unstimulated PBMC proliferate well after subsequent stimulation with anti-CD3, whereas IL-2 induces only limited proliferation. In contrast, preactivated cells respond only minimally to restimulation with anti-CD3, but IL-2 induces a marked proliferation. Both preactivated and unstimulated Tc respond well to restimulation by phytohaemagglutinin (PHA). In contrast, preactivated Tc show only a weak response to concanavalin A. Interestingly, when cells have been allowed to rest for 168 h, the responsiveness of preactivated Tc is restored. Immunoblots reveal that preactivated cells have a higher intracellular content of zeta-chain and p56lck. No differences are found concerning apoptosis after restimulation with anti-CD3 or the expression of ERK 1/2. The unresponsiveness to restimulation is due to an impairment of the transcription of the IL-2 gene and this defect is temporary. Despite the lack of proliferation, preactivated Tc phenotypically maintain an intermediate stage of activation. These data show how the same cell population can change its functional phenotype into a non-responder state.
Collapse
Affiliation(s)
- M D Köller
- Department Internal Medicine III, Division of Rheumatology, University of Vienna, Austria.
| | | | | | | | | | | | | |
Collapse
|
76
|
Huang CT, Huso DL, Lu Z, Wang T, Zhou G, Kennedy EP, Drake CG, Morgan DJ, Sherman LA, Higgins AD, Pardoll DM, Adler AJ. CD4+ T cells pass through an effector phase during the process of in vivo tolerance induction. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:3945-53. [PMID: 12682221 DOI: 10.4049/jimmunol.170.8.3945] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
An important process in the generation of tolerance to peripheral self-Ags is the induction of unresponsiveness in mature specific T cells. Although the end stage of this process, termed anergy, is well defined, the pathway by which naive T cells become anergic remains to be elucidated. Using an in vivo self-tolerance model, we demonstrate that CD4(+) T cells pass through a significant effector stage on their way to an anergic state. This stage is characterized by production of effector cytokines, provision of help for CD8(+) T cells, and induction of in vivo pathology within organs that express cognate Ag. These results suggest that the initial activation stage in T cell tolerance is similar to that seen in memory induction. They also suggest that autoimmune pathology can result during the natural process of tolerance induction rather than requiring that tolerance be broken.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Autoantigens/biosynthesis
- Autoimmune Diseases/genetics
- Autoimmune Diseases/mortality
- Autoimmune Diseases/pathology
- CD4-Positive T-Lymphocytes/cytology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/transplantation
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Clonal Anergy/genetics
- Epitopes, T-Lymphocyte/biosynthesis
- Epitopes, T-Lymphocyte/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/biosynthesis
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Histocompatibility Antigens Class II/genetics
- Histocompatibility Antigens Class II/immunology
- Immune Tolerance/genetics
- Lung Diseases/genetics
- Lung Diseases/immunology
- Lung Diseases/mortality
- Lung Diseases/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mice, Transgenic
- Models, Immunological
- Rats
- Receptors, Antigen, T-Cell/biosynthesis
- Receptors, Antigen, T-Cell/genetics
- Self Tolerance/genetics
- Self Tolerance/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/transplantation
Collapse
Affiliation(s)
- Ching-Tai Huang
- Oncology Center and Division of Comparative Medicine, Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Martinic MM, Rülicke T, Althage A, Odermatt B, Höchli M, Lamarre A, Dumrese T, Speiser DE, Kyburz D, Hengartner H, Zinkernagel RM. Efficient T cell repertoire selection in tetraparental chimeric mice independent of thymic epithelial MHC. Proc Natl Acad Sci U S A 2003; 100:1861-6. [PMID: 12574503 PMCID: PMC149924 DOI: 10.1073/pnas.252641399] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Nonthymic epithelial cells were compared with thymic epithelial cells for their role in T cell repertoire selection. Tetraparental aggregation chimeras were generated from T and B cell-deficient mice (H-2(d) SCID or H-2(b) Rag-/-) and thymus-deficient nude mice (H-2(b) or H-2(d)). These tetraparental mice showed primary protective CD8(+) T cell responses, after lymphocytic choriomeningitis virus infection, that were peptide-specifically restricted to either thymic or nonthymic epithelial MHC at comparable levels. These chimeras also mounted neutralizing IgG responses dependent on cognate CD4(+) T helper cell activity restricted to nonthymic epithelial MHC. Therefore, in contrast to earlier results with irradiation or thymus chimeras, these relatively undisturbed tetraparental mice reveal that the MHC of nonthymic epithelial cells efficiently selects a functional T cell repertoire.
Collapse
Affiliation(s)
- Marianne M Martinic
- Institute of Experimental Immunology, Department of Pathology, University Hospital Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Celis E. Getting peptide vaccines to work: just a matter of quality control? J Clin Invest 2003. [PMID: 12488425 DOI: 10.1172/jci200217405] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Esteban Celis
- Department of Immunology, Mayo Clinic, Rochester, Minnesota 55905, USA.
| |
Collapse
|
79
|
Salomone T, Tosi P, Raiti C, Guariento A, Tomassetti P, Migliori M, Saieva C, Romboli M, Gullo L. Apoptosis in the peripheral blood mononuclear cells as a self-limitation process in human acute pancreatitis. Pancreatology 2003; 2:204-10. [PMID: 12138725 DOI: 10.1159/000058034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND/AIM Acute pancreatitis is primed and sustained by a chain of immuno-inflammatory factors. In this study, we investigated the possible existence of peripheral blood mononuclear cell apoptosis as a self-limitation mechanism in acute pancreatitis. METHODS Peripheral blood mononuclear cell apoptosis was determined cytofluorometrically daily for 10 days from the onset of the illness in 27 consecutive patients (18 having mild uncomplicated acute pancreatitis and 9 having pancreatitis with complications) and was related to peripheral blood counts, including reticulocytes and reticulocyte fractions, and albumin, fibrinogen, and C-reactive protein levels. RESULTS In the 18 patients with uncomplicated acute pancreatitis, the rate of peripheral blood apoptosis increased progressively until days 5-6 and then decreased. The 9 patients who developed complications showed levels of peripheral blood apoptosis stable across the five periods and lower than those with uncomplicated disease during the first four periods. This difference was statistically significant (p = 0.002) only on days 7-8. On days 9-10, the patients with complications showed higher levels of peripheral blood apoptosis than those with mild uncomplicated acute pancreatitis (p = 0.0005). Peripheral blood apoptosis was not significantly related to the other laboratory parameters, but there was a trend towards an inverse relation to reticulocytes and total leucocytes (p < 0.09). CONCLUSIONS Peripheral blood apoptosis may act as a mechanism of self-limitation of the process of acute pancreatitis. Its effects, however, seem to be hampered and delayed by the presence of complications.
Collapse
|
80
|
Affiliation(s)
- Esteban Celis
- Department of Immunology, Mayo Clinic, Rochester, Minnesota 55905, USA.
| |
Collapse
|
81
|
Nopora A, Brocker T. Bcl-2 controls dendritic cell longevity in vivo. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:3006-14. [PMID: 12218115 DOI: 10.4049/jimmunol.169.6.3006] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Dendritic cells (DC) were found to down-regulate Bcl-2 protein upon maturation in vivo. Because Bcl-2 has been shown to exert anti-apoptotic functions, down-regulation of Bcl-2 could be a mechanism by which DC longevity is controlled. To dysregulate this potential control system and to study the role of Bcl-2 in DC, we expressed human Bcl-2 under control of the murine CD11c-promoter as a transgene specifically in DC and show that DC frequencies and numbers increase in transgenic mice. In vivo bromodeoxyuridin, as well as adoptive, DC transfer studies show that the relative turnover/survival of mature Bcl-2 transgenic DC is increased. This had a direct impact on CD4+ T cell, as well as humoral immune, responses, which were elevated in transgenic animals. When Bcl-2 transgenic DC were used as DC vaccines, they induced 2- to 3-fold greater expansion of Ag-specific CTL, and stronger in vivo cytotoxicity. Overall, these data indicate that down-regulation of Bcl-2 controls DC longevity, which in turn directly regulates immune responses and the efficacy of DC when used as vaccines.
Collapse
Affiliation(s)
- Adam Nopora
- Institute for Immunology, Ludwig-Maximilians University, Munich, Germany
| | | |
Collapse
|
82
|
Nguyen LT, Bachmann MF, Ohashi PS. Contribution of LCMV transgenic models to understanding T lymphocyte development, activation, tolerance, and autoimmunity. Curr Top Microbiol Immunol 2002; 263:119-43. [PMID: 11987812 DOI: 10.1007/978-3-642-56055-2_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- L T Nguyen
- Department of Immunology, Ontario Cancer Institute, 610 University Avenue, Toronto, Ontario, Canada, M5G 2M9
| | | | | |
Collapse
|
83
|
Unsoeld H, Krautwald S, Voehringer D, Kunzendorf U, Pircher H. Cutting edge: CCR7+ and CCR7- memory T cells do not differ in immediate effector cell function. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:638-41. [PMID: 12097363 DOI: 10.4049/jimmunol.169.2.638] [Citation(s) in RCA: 151] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
It has been proposed that expression of the chemokine receptor CCR7 represents a defining factor for nonpolarized central (CCR7(+)) and polarized effector memory (CCR7(-)) T cells. In this study, we have tested this hypothesis using in vivo-activated T cells from P14 and SMARTA TCR-transgenic (tg) mice specific for MHC class I- and II-restricted epitopes of the lymphocytic choriomeningitis virus (LCMV) glycoprotein. CCR7 cell surface expression on TCR-tg cells was monitored with a CC chemokine ligand 19-Ig fusion protein. CC chemokine ligand 19-Ig staining separated TCR-tg cells activated by LCMV infection into CCR7(-) and CCR7(+) effector/memory T cell populations. Nonetheless, both T cell populations isolated from spleen and liver produced identical amounts of IFN-gamma after short-term Ag stimulation. Furthermore, CCR7(+) and CCR7(-) CD8 TCR-tg cells from LCMV-infected mice exhibited similar lytic activity against LCMV peptide-coated target cells. These results question the proposed concept of differential effector cell function of CCR7(+) and CCR7(-) memory T cells.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/virology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/virology
- Cell Membrane/immunology
- Cell Membrane/metabolism
- Cytokines/biosynthesis
- Cytotoxicity, Immunologic/genetics
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Glycoproteins/genetics
- Glycoproteins/immunology
- Immunologic Memory/genetics
- Kinetics
- Lymphocyte Activation/genetics
- Lymphocytic choriomeningitis virus/genetics
- Lymphocytic choriomeningitis virus/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Molecular Sequence Data
- Peptide Fragments/genetics
- Peptide Fragments/immunology
- Receptors, CCR7
- Receptors, Chemokine/biosynthesis
- Receptors, Chemokine/deficiency
- Receptors, Chemokine/genetics
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/virology
- Viral Proteins/genetics
- Viral Proteins/immunology
Collapse
Affiliation(s)
- Heike Unsoeld
- Department of Immunology, Institute for Medical Microbiology and Hygiene, University of Freiburg, Freiburg, Germany
| | | | | | | | | |
Collapse
|
84
|
Abstract
Memory T cells differ from naive T cells in that they respond more rapidly and in greater numbers. In addition, memory T cells are generally believed to be less susceptible to tolerance induction than naive T cells. In this study, we show that this is not the case. Using two different methods of tolerance induction, peptide-induced tolerance and crosstolerance, we present evidence that memory CD8(+) T cells are as susceptible to tolerance as naive cells. These results have a direct impact on manipulating T cell responses to self-antigens in order to improve immunotherapy of cancer and autoimmune diseases.
Collapse
Affiliation(s)
- Huub T C Kreuwel
- Department of Immunology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
85
|
Schmidt CS, Mescher MF. Peptide antigen priming of naive, but not memory, CD8 T cells requires a third signal that can be provided by IL-12. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:5521-9. [PMID: 12023347 DOI: 10.4049/jimmunol.168.11.5521] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Challenge with peptide Ag in the absence of adjuvant results in tolerance of CD8 T cells specific for the Ag. In contrast, administration of IL-12 along with peptide results in massive clonal expansion, development of effector function, and establishment of a long-lived memory population. Using adoptive transfer of TCR-transgenic CD8 T cells, this effect of IL-12 is shown to be independent of CD4 T cells and to require costimulation provided by CD28 and possibly LFA-1. IL-12 supports responses when IL-12Rbeta1-deficient mice are used as recipients for the adoptively transferred CD8 T cells, demonstrating that the IL-12 is acting directly on the T cells rather than on host APC. These results provide strong support for a three-signal model for in vivo activation of naive CD8 T cells by peptide Ag, in which the presence or absence of the third signal determines whether tolerance or activation occurs. In contrast, memory CD8 T cells are effectively activated by peptide Ag in the absence of IL-12 or adjuvant.
Collapse
Affiliation(s)
- Clint S Schmidt
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | | |
Collapse
|
86
|
Zarei S, Leuba F, Arrighi JF, Hauser C, Piguet V. Transduction of dendritic cells by antigen-encoding lentiviral vectors permits antigen processing and MHC class I-dependent presentation. J Allergy Clin Immunol 2002; 109:988-94. [PMID: 12063529 DOI: 10.1067/mai.2002.124663] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Because antigen-presenting dendritic cells (DCs) play a major role in the polarization of T cells, including T(H)2 cells involved in allergy, strategies to modify DCs genetically are required. OBJECTIVE The purpose of this investigation was to transduce murine bone marrow-derived DCs with lentiviral vectors encoding antigen to demonstrate antigen processing and MHC class I-dependent presentation. METHODS Bone marrow leukocytes were incubated with antigen-encoding lentiviral constructs and cultured with GM-CSF, IL-4, and Flt-3 ligand. The capacity of the resulting DCs to express, process, and present antigen was tested in vitro. RESULTS An average of 40% of DCs expressed antigen after 1 week of culture when antigen encoded by the lentiviral vector construct was green fluorescent protein. To demonstrate that transduced antigen can be presented by DCs on MHC class I, we chose the lymphocytic choriomeningitis virus glycoprotein (gp) as a model antigen, inasmuch as it is recognized by CD8 T cells from transgenic mice expressing an MHC class I-restricted T-cell receptor specific for the epitope of positions 33 through 41 of gp. DCs transduced with lentiviral construct encoding gp and matured with LPS activated transgenic T cells in an antigen-specific fashion. Using transporter associated with antigen presentation (TAP)-deficient mice, we show that presentation of the gp33-41 epitope is TAP-dependent, confirming processing of gp by the endogenous pathway. CONCLUSIONS These results demonstrate that CD8 T cells can recognize MHC class I epitopes processed from antigen in DCs transduced with lentiviral vectors. Lentiviral transduction of DCs and antigen presentation to CD8 T cells could be exploited for immunotherapy, because allergen-specific CD8 T cells have been shown to be suppressive in IgE-dependent allergy models.
Collapse
Affiliation(s)
- Shohreh Zarei
- Division of Allergy and Immunology and the Division of Hematology, Department of Dermatology, University Hospital of Geneva, Switzerland
| | | | | | | | | |
Collapse
|
87
|
Roth E, Schwartzkopff J, Pircher H. CD40 ligation in the presence of self-reactive CD8 T cells leads to severe immunopathology. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:5124-9. [PMID: 11994466 DOI: 10.4049/jimmunol.168.10.5124] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Previous work has shown that stimulation of APCs via CD40 strongly influences the outcome of a CD8 T cell response. In this study, we examined the effect of CD40 ligation on peripheral tolerance induction of self-reactive CD8 T cells in an adoptive transfer model. Naive CD8 T cells from TCR-transgenic (tg) mice specific for the gp33 epitope of lymphocytic choriomeningitis virus were tolerized when transferred into H8-tg mice expressing the gp33 epitope under the control of a MHC class I promoter. However, if the H8 recipient mice were treated with agonistic anti-CD40 Abs, TCR-tg cells vigorously proliferated, and induced destruction of lymphoid organs and hepatitis. Break of peripheral tolerance induction was B cell independent and did not require CD28/B7 interactions. These findings provide further in vivo evidence for the crucial role of the activation state of the APC in peripheral tolerance induction and suggest the need for caution in systemically activating APC via CD40 ligation in the presence of self-reactive T cells.
Collapse
Affiliation(s)
- Evelyn Roth
- Institute for Medical Microbiology and Hygiene, Department of Immunology, University of Freiburg, Hermann-Herder-Strasse 11, D-79104 Freiburg, Germany
| | | | | |
Collapse
|
88
|
Brassard DL, Grace MJ, Bordens RW. Interferon‐α as an immunotherapeutic protein. J Leukoc Biol 2002. [DOI: 10.1189/jlb.71.4.565] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
Affiliation(s)
- Diana L. Brassard
- Bioanalytical Development, Schering‐Plough Research Institute, Union, New Jersey
| | - Michael J. Grace
- Bioanalytical Development, Schering‐Plough Research Institute, Union, New Jersey
| | - Ronald W. Bordens
- Bioanalytical Development, Schering‐Plough Research Institute, Union, New Jersey
| |
Collapse
|
89
|
Nguyen LT, Elford AR, Murakami K, Garza KM, Schoenberger SP, Odermatt B, Speiser DE, Ohashi PS. Tumor growth enhances cross-presentation leading to limited T cell activation without tolerance. J Exp Med 2002; 195:423-35. [PMID: 11854356 PMCID: PMC2193619 DOI: 10.1084/jem.20010032] [Citation(s) in RCA: 110] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Using a tumor model of spontaneously arising insulinomas expressing a defined tumor-associated antigen, we investigated whether tumor growth promotes cross-presentation and tolerance of tumor-specific T cells. We found that an advanced tumor burden enhanced cross-presentation of tumor-associated antigens to high avidity tumor-specific T cells, inducing T cell proliferation and limited effector function in vivo. However, contrary to other models, tumor-specific T cells were not tolerized despite a high tumor burden. In fact, in tumor-bearing mice, persistence and responsiveness of adoptively transferred tumor-specific T cells were enhanced. Accordingly, a potent T cell-mediated antitumor response could be elicited by intravenous administration of tumor-derived peptide and agonistic anti-CD40 antibody or viral immunization and reimmunization. Thus, in this model, tumor growth promotes activation of high avidity tumor-specific T cells instead of tolerance. Therefore, the host remains responsive to T cell immunotherapy.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antibodies, Monoclonal/immunology
- Antigen Presentation
- Antigens, Neoplasm/immunology
- Antigens, Tumor-Associated, Carbohydrate/administration & dosage
- Antigens, Tumor-Associated, Carbohydrate/immunology
- CD40 Antigens/immunology
- Cell Division
- Flow Cytometry
- Hyaluronan Receptors/immunology
- Hyaluronan Receptors/metabolism
- Hypoglycemia/complications
- Immune Tolerance
- Immunologic Surveillance
- Immunotherapy, Active
- Insulinoma/complications
- Insulinoma/immunology
- Insulinoma/pathology
- Insulinoma/therapy
- Lymph Nodes/immunology
- Lymphocyte Activation
- Mice
- Mice, Transgenic
- Radiation Chimera
- Survival Analysis
- T-Lymphocytes, Cytotoxic/cytology
- T-Lymphocytes, Cytotoxic/immunology
- Time Factors
Collapse
Affiliation(s)
- Linh T Nguyen
- Departments of Immunology and Medical Biophysics, Ontario Cancer Institute, 610 University Ave., Toronto, Ontario M5G 2M9, Canada
| | | | | | | | | | | | | | | |
Collapse
|
90
|
Soruri A, Schweyer S, Radzun HJ, Fayyazi A. Mycobacterial antigens induce apoptosis in human purified protein derivative-specific alphabeta T lymphocytes in a concentration-dependent manner. Immunology 2002; 105:222-30. [PMID: 11872098 PMCID: PMC1782647 DOI: 10.1046/j.0019-2805.2001.01355.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The morbidity and lethality of tuberculosis is partially the result of an ineffective delayed-type hypersensitivity reaction which causes caseating granulomas in the lung and other organs. Recently we showed that during caseation besides macrophages numerous Fas+ FasL+ lymphocytes undergo apoptosis and postulated that this phenomenon may be due to activation-induced cell death (AICD) as a consequence of T-lymphocyte reactivation via bacillary antigens. As purified protein derivative of Mycobacterium tuberculosis (Mtb-PPD) provokes caseation in tuberculosis patients, the question arose as to whether bacillary antigens are responsible for AICD within caseous areas. In the present study Mtb-PPD-specific T helper 1 (Th1)-differentiated T lymphocytes were generated in vitro. Reactivation of these cells with Mtb-PPD resulted in a concentration-dependent hyporesponsiveness, which was due to an increase in apoptosis of gammadelta+, alphabeta+ CD4+ as well as alphabeta+ CD8+ T lymphocytes as assessed by the demonstration of the apoptosis-associated mitochondrial membrane protein 7A6 and DNA fragmentation. Blocking experiments demonstrated that Mtb-PPD antigens exploited the Fas/FasL system to induce apoptosis in Mtb-PPD-specific T lymphocytes. These results may support the hypothesis that in tubercle granulomas with caseation T lymphocytes undergo AICD following reactivation by bacillary antigens, thus contributing to the persistence of tuberculosis.
Collapse
Affiliation(s)
- Afsaneh Soruri
- Department of Immunology, Georg August University Goettingen, Robert-Koch-Strasse 40, D-37075 Goettingen, Germany
| | | | | | | |
Collapse
|
91
|
Peakman M, Dayan CM. Antigen-specific immunotherapy for autoimmune disease: fighting fire with fire? Immunology 2001; 104:361-6. [PMID: 11899420 PMCID: PMC1783327 DOI: 10.1046/j.1365-2567.2001.01335.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- M Peakman
- Department of Immunology, Guy's, King's and St Thomas' School of Medicine, Rayne Institute, 123 Coldharbour Lane, London SE5 9NU, UK.
| | | |
Collapse
|
92
|
Voehringer D, Blaser C, Brawand P, Raulet DH, Hanke T, Pircher H. Viral infections induce abundant numbers of senescent CD8 T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:4838-43. [PMID: 11673487 DOI: 10.4049/jimmunol.167.9.4838] [Citation(s) in RCA: 191] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Viral infections are often accompanied by extensive proliferation of reactive CD8 T cells. After a defined number of divisions, normal somatic cells enter a nonreplicative stage termed senescence. In the present study we have identified the inhibitory killer cell lectin-like receptor G1 (KLRG1) as a unique marker for replicative senescence of murine CD8 T cells. KLRG1 expression was induced in a substantial portion (30-60%) of CD8 T cells in C57BL/6 mice infected with lymphocytic choriomeningitis virus (LCMV), vesicular stomatitis virus, or vaccinia virus. Similarly, KLRG1 was found on a large fraction of LCMV gp33 peptide-specific TCR-transgenic (tg) effector and memory cells activated in vivo using an adoptive transfer model. Transfer experiments with CFSE-labeled TCR-tg cells into LCMV-infected hosts further indicated that induction of KLRG1 expression required an extensive number of cell divisions. Most importantly, KLRG1(+) TCR-tg effector/memory cells could efficiently lyse target cells and secrete cytokines, but were severely impaired in their ability to proliferate after Ag stimulation. Thus, this study demonstrates that senescent CD8 T cells are induced in abundant numbers during viral infections in vivo.
Collapse
Affiliation(s)
- D Voehringer
- Institute for Medical Microbiology and Hygiene, Department of Immunology, University of Freiburg, Freiburg, Germany
| | | | | | | | | | | |
Collapse
|
93
|
Shi W, Liu J, Huang Y, Qiao L. Papillomavirus pseudovirus: a novel vaccine to induce mucosal and systemic cytotoxic T-lymphocyte responses. J Virol 2001; 75:10139-48. [PMID: 11581382 PMCID: PMC114588 DOI: 10.1128/jvi.75.21.10139-10148.2001] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Intestinal mucosa is a portal for many infectious pathogens. Systemic immunization, in general, does not induce a cytotoxic T-lymphocyte (CTL) response at the mucosal surface. Because papillomavirus (PV) naturally infects mucosa and skin, we determined whether PV pseudovirus, i.e., PV-like particles in which unrelated DNA plasmids are packaged, could generate specific mucosal immunity. We found that the pseudovirus that encoded the lymphocytic choriomeningitis virus gp33 epitope induced a stronger CTL response than a DNA vaccine (plasmid) encoding the same epitope given systemically. The virus-like particles that were used to make the pseudoviruses provided an adjuvant effect for induction of CTLs by the DNA vaccine. The PV pseudovirus pseudoinfected mucosal and systemic lymphoid tissues when administered orally. Oral immunization with the pseudovirus encoding human PV type 16 mutant E7 induced mucosal and systemic CTL responses. In comparison, a DNA vaccine encoding E7, when given orally, did not induce a CTL response in intestinal mucosal lymphoid tissue. Further, oral immunization with the human PV pseudovirus encoding E7 protected mice against mucosal challenge with an E7-expressing bovine PV pseudovirus. Thus, PV pseudovirus can be used as a novel vaccine to induce mucosal and systemic CTL responses.
Collapse
Affiliation(s)
- W Shi
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois 60153, USA
| | | | | | | |
Collapse
|
94
|
Abstract
Tolerance is a state in which the immune system as a whole fails to make an active response to antigen. Three mutually exclusive mechanisms appear to account for the fate of antigen-specific peripheral T cells within a tolerant animal: maintenance of naive status, deletion after responding to antigen, and long term survival after responding to antigen, a mechanism that should probably be considered part of the spectrum of memory responses. The types and functional status of the DCs that present antigen in each case remain controversial. This review will summarize the indirect evidence that underlies some of the hypotheses that account for peripheral T cell tolerance.
Collapse
Affiliation(s)
- B Fazekas de St Groth
- Centenary Institute of Cancer Medicine and Cell Biology, Locked Bag No. 6, Newtown, NSW, 2042, Australia.
| |
Collapse
|
95
|
Lechler R, Chai JG, Marelli-Berg F, Lombardi G. T-cell anergy and peripheral T-cell tolerance. Philos Trans R Soc Lond B Biol Sci 2001; 356:625-37. [PMID: 11375066 PMCID: PMC1088450 DOI: 10.1098/rstb.2001.0844] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The discovery that T-cell recognition of antigen can have distinct outcomes has advanced understanding of peripheral T-cell tolerance, and opened up new possibilities in immunotherapy. Anergy is one such outcome, and results from partial T-cell activation. This can arise either due to subtle alteration of the antigen, leading to a lower-affinity cognate interaction, or due to a lack of adequate co-stimulation. The signalling defects in anergic T cells are partially defined, and suggest that T-cell receptor (TCR) proximal, as well as downstream defects negatively regulate the anergic T cell's ability to be activated. Most importantly, the use of TCR-transgenic mice has provided compelling evidence that anergy is an in vivo phenomenon, and not merely an in vitro artefact. These findings raise the question as to whether anergic T cells have any biological function. Studies in rodents and in man suggest that anergic T cells acquire regulatory properties; the regulatory effects of anergic T cells require cell to cell contact, and appear to be mediated by inhibition of antigen-presenting cell immunogenicity. Close similarities exist between anergic T cells, and the recently defined CD4+ CD25+ population of spontaneously arising regulatory cells that serve to inhibit autoimmunity in mice. Taken together, these findings suggest that a spectrum of regulatory T cells exists. At one end of the spectrum are cells, such as anergic and CD4+ CD25+ T cells, which regulate via cell-to-cell contact. At the other end of the spectrum are cells which secrete antiinflammatory cytokines such as interleukin 10 and transforming growth factor-beta. The challenge is to devise strategies that reliably induce T-cell anergy in vivo, as a means of inhibiting immunity to allo- and autoantigens.
Collapse
Affiliation(s)
- R Lechler
- Department of Immunology, Imperial College School of Medicine, Hammersmith Campus, Du Cane Road, London W12 0NN,UK.
| | | | | | | |
Collapse
|
96
|
Wang B, Norbury CC, Greenwood R, Bennink JR, Yewdell JW, Frelinger JA. Multiple paths for activation of naive CD8+ T cells: CD4-independent help. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:1283-9. [PMID: 11466344 DOI: 10.4049/jimmunol.167.3.1283] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
CD8(+) CTLs play a pivotal role in immune responses against many viruses and tumors. Two models have been proposed. The "three-cell" model focuses on the role of CD4(+) T cells, proposing that help is only provided to CTLs by CD4(+) T cells that recognize Ag on the same APC. The sequential "two-cell" model proposes that CD4(+) T cells can first interact with APCs, which in turn activate naive CTLs. Although these models provide a general framework for the role of CD4(+) T cells in mediating help for CTLs, a number of issues are unresolved. We have investigated the induction of CTL responses using dendritic cells (DCs) to immunize mice against defined peptide Ags. We find that help is required for activation of naive CTLs when DCs are used as APCs, regardless of the origin or MHC class I restriction of the peptides we studied in this system. However, CD8(+) T cells can provide self-help if they are present at a sufficiently high precursor frequency. The important variable is the total number of T cells responding, because class II-knockout DCs pulsed with two noncompeting peptides are effective in priming.
Collapse
Affiliation(s)
- B Wang
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | | | | | |
Collapse
|
97
|
Lechler R, Chai JG, Marelli-Berg F, Lombardi G. The contributions of T-cell anergy to peripheral T-cell tolerance. Immunology 2001; 103:262-9. [PMID: 11454055 PMCID: PMC1783252 DOI: 10.1046/j.1365-2567.2001.01250.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- R Lechler
- Department of Immunology, Imperial College School of Medicine, London, UK.
| | | | | | | |
Collapse
|
98
|
Bu P, Keshavarzian A, Stone DD, Liu J, Le PT, Fisher S, Qiao L. Apoptosis: one of the mechanisms that maintains unresponsiveness of the intestinal mucosal immune system. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:6399-403. [PMID: 11342665 DOI: 10.4049/jimmunol.166.10.6399] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Intestinal mucosa is constantly exposed to environmental AGS: Activation of lamina propria (LP) T cells by luminal Ags may lead to the production of inflammatory cytokines and subsequent mucosal inflammation and tissue damage. However, in normal circumstances, LP T cells do not respond to antigenic stimulation. The mechanisms of this unresponsiveness in healthy subjects are not fully understood. In this study, we found by in vivo analysis that, except for T cells in lymph nodules of the mucosa, 15% of LP T cells underwent apoptosis in normal individuals. In contrast, there was a marked reduction in apoptosis of LP T cells in patients with inflammatory bowel disease (Crohn's disease and ulcerative colitis) and those with specific colitis. Our findings suggest that apoptosis might be a mechanism that turns off mucosal T cell responses to environmental Ags in healthy subjects, and resistance to apoptosis could be an important cause of mucosal immune dysregulation and tissue inflammation in colitis.
Collapse
Affiliation(s)
- P Bu
- Department of Microbiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | | | | | | | | | | | | |
Collapse
|
99
|
Ludewig B, McCoy K, Pericin M, Ochsenbein AF, Dumrese T, Odermatt B, Toes RE, Melief CJ, Hengartner H, Zinkernagel RM. Rapid peptide turnover and inefficient presentation of exogenous antigen critically limit the activation of self-reactive CTL by dendritic cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:3678-87. [PMID: 11238607 DOI: 10.4049/jimmunol.166.6.3678] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
This study evaluated to what extent presentation of exogenously acquired self-Ags via MHC class I molecules on DC might contribute to the activation of self-reactive CTL and subsequent development of autoimmune disease. We show here by using the rat insulin promotor lymphocytic choriomeningitis virus glycoprotein model of autoimmune diabetes that the activation of self-reactive CTL by DC after uptake of exogenous Ag is very limited, first by the short half-life of MHC class I-associated peptides on DC in vitro and in vivo, and second by the rather inefficient MHC class I presentation of cell-associated self-Ags by DC. These two mechanisms are probably crucial in establishing high thresholds for the induction of self-reactive CTL that prevent autoimmune sequelae after release of sequestered and previously immunologically ignored tissue Ags.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigen Presentation/genetics
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Antigens, Viral/metabolism
- Cytotoxicity, Immunologic/genetics
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/transplantation
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/immunology
- Diabetes Mellitus, Experimental/pathology
- Glycoproteins/immunology
- Glycoproteins/metabolism
- Histocompatibility Antigens Class I/immunology
- Histocompatibility Antigens Class I/metabolism
- Injections, Subcutaneous
- Insulin/genetics
- Islets of Langerhans/immunology
- Islets of Langerhans/pathology
- Lymphocyte Activation/genetics
- Lymphocytic choriomeningitis virus/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Molecular Sequence Data
- Peptide Fragments/immunology
- Peptide Fragments/metabolism
- Peptides/immunology
- Peptides/metabolism
- Promoter Regions, Genetic/genetics
- Promoter Regions, Genetic/immunology
- Rats
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Helper-Inducer/immunology
- Tumor Cells, Cultured/transplantation
- Viral Proteins
Collapse
Affiliation(s)
- B Ludewig
- Institute of Experimental Immunology, Department of Pathology, University of Zürich, Zürich, Switzerland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Affiliation(s)
- R M Welsh
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| |
Collapse
|