Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 6(1): e14502, 2011 Jan 06.
Article in English | MEDLINE | ID: mdl-21253594

ABSTRACT

BACKGROUND: Memory CD8 T cells form an essential part of protective immunity against viral infections. Antigenic load, costimulation, CD4-help, cytokines and chemokines fluctuate during the course of an antiviral immune response thus affecting CD8 T cell activation and memory conversion. METHODOLOGY/PRINCIPAL FINDINGS: In the present study, naïve TCR transgenic LCMV-specific P14 CD8 T cells engaged at a late stage during the acute antiviral LCMV response showed reduced expansion kinetics but greater memory conversion in the spleen. Such late activated cells displayed a memory precursor effector phenotype already at the peak of the systemic antiviral response, suggesting that the environment determined their fate during antigen encounter. In the spleen, the majority of late transferred cells exhibited a central memory phenotype compared to the effector memory displayed by the early transferred cells. Increasing the inflammatory response by exogenous administration of IFNγ, PolyI:C or CpG did not affect memory conversion in the late transferred group, suggesting that the diverging antigen load early versus later during acute infection had determined their fate. In agreement, reduction in the LCMV antigenic load after ribavirin treatment enhanced the contribution of early transferred cells to the long lasting memory pool. CONCLUSIONS/SIGNIFICANCE: Our results show that naïve CD8 cells, exposed to reduced duration or concentration of antigen during viral infection convert into memory more efficiently, an observation that could have significant implications for vaccine design.


Subject(s)
Antigens, Viral/analysis , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Virus Diseases/immunology , Animals , Antigens, Viral/immunology , Cells, Cultured , Immunity , Inflammation , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , T-Cell Antigen Receptor Specificity/immunology , Time Factors
2.
Diabetes ; 57(10): 2684-92, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18689691

ABSTRACT

OBJECTIVE: Transforming growth factor-beta (TGF-beta) can exhibit strong immune suppression but has also been shown to promote T-cell growth. We investigated the differential effect of this cytokine on CD8(+) T-cells in autoimmunity and antiviral immunity. RESEARCH DESIGN AND METHODS: We used mouse models for virally induced type 1 diabetes in conjunction with transgenic systems enabling manipulation of TGF-beta expression or signaling in vivo. RESULTS: Surprisingly, when expressed selectively in the pancreas, TGF-beta reduced apoptosis of differentiated autoreactive CD8(+) T-cells, favoring their expansion and infiltration of the islets. These results pointed to drastically opposite roles of TGF-beta on naïve compared with antigen-experienced/memory CD8(+) T-cells. Indeed, in the absence of functional TGF-beta signaling in T-cells, fast-onset type 1 diabetes caused by activation of naïve CD8(+) T-cells occurred faster, whereas slow-onset disease depending on accumulation and activation of antigen-experienced/memory CD8(+) T-cells was decreased. TGF-beta receptor-deficient CD8(+) T-cells showed enhanced activation and expansion after lymphocytic choriomeningitis virus infection in vivo but were more prone to apoptosis once antigen experienced and failed to survive as functional memory cells. In vitro, TGF-beta suppressed naïve CD8(+) T-cell activation and gamma-interferon production, whereas memory CD8(+) T-cells stimulated in the presence of TGF-beta showed enhanced survival and increased production of interleukin-17 in conjunction with gamma-interferon. CONCLUSIONS: The effect of TGF-beta on CD8(+) T-cells is dependent on their differentiation status and activation history. These results highlight a novel aspect of the pleiotropic nature of TGF-beta and have implications for the design of immune therapies involving this cytokine.


Subject(s)
Apoptosis/drug effects , CD8-Positive T-Lymphocytes/drug effects , Transforming Growth Factor beta/pharmacology , Animals , Apoptosis/immunology , Autoimmunity/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Survival/drug effects , Cell Survival/immunology , Cells, Cultured , Flow Cytometry , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Pancreas/drug effects , Pancreas/immunology , Pancreas/metabolism
3.
Diabetes ; 56(4): 1059-68, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17395746

ABSTRACT

During an autoimmune process, the autoaggressive response spreads from the initiating autoantigen to other antigens expressed in the target organ. Based on evidence from experimental models for multiple sclerosis, such "antigenic spreading" can play an important role in the exacerbation of clinical disease. We evaluated whether pathogenesis of spontaneous diabetes in NOD mice could be accelerated in a similar way when a novel autoantigen was expressed in pancreatic beta-cells. Unexpectedly, we found that the expression of the lymphocytic choriomeningitis virus nucleoprotein only led to marginal enhancement of diabetes, although such NOD-nucleoprotein mice were not tolerant to nucleoprotein. Although the frequency of nucleoprotein-specific CD8 T-cells in the pancreatic draining lymph node was comparable with the frequency of islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP)-specific T-cells, more IGRP-specific CD8 T-cells were found both systemically and in the islets where there was a fourfold increase. Interestingly, and in contrast to nucleoprotein-specific CD8 T-cells, IGRP-specific T-cells showed increased CXCR3 expression. Thus, autoreactivity toward de novo-expressed beta-cell autoantigens will not accelerate autoimmunity unless large numbers of antigen-experienced autoreactive T-cells expressing the appropriate chemokine receptors are present.


Subject(s)
Autoantigens/immunology , Diabetes Mellitus, Type 1/physiopathology , Insulin-Secreting Cells/immunology , Animals , Cloning, Molecular , DNA Primers , Diabetes Mellitus, Type 1/immunology , Enzyme-Linked Immunosorbent Assay , Insulin/analysis , Lymphocytic choriomeningitis virus/genetics , Mice , Mice, Inbred NOD , Mice, Transgenic , Nucleoproteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Viral Proteins/genetics
4.
J Exp Med ; 203(11): 2461-72, 2006 Oct 30.
Article in English | MEDLINE | ID: mdl-17030951

ABSTRACT

A defining characteristic of persistent viral infections is the loss and functional inactivation of antiviral effector T cells, which prevents viral clearance. Interleukin-10 (IL-10) suppresses cellular immune responses by modulating the function of T cells and antigen-presenting cells. In this paper, we report that IL-10 production is drastically increased in mice persistently infected with lymphocytic choriomeningitis virus. In vivo blockade of the IL-10 receptor (IL-10R) with a neutralizing antibody resulted in rapid resolution of the persistent infection. IL-10 secretion was diminished and interferon gamma production by antiviral CD8+ T cells was enhanced. In persistently infected mice, CD8alpha+ dendritic cell (DC) numbers declined early after infection, whereas CD8alpha- DC numbers were not affected. CD8alpha- DCs supported IL-10 production and subsequent dampening of antiviral T cell responses. Therapeutic IL-10R blockade broke the cycle of IL-10-mediated immune suppression, preventing IL-10 priming by CD8alpha- DCs and enhancing antiviral responses and thereby resolving infection without causing immunopathology.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/therapy , Lymphocytic choriomeningitis virus , Receptors, Interleukin-10/antagonists & inhibitors , Receptors, Interleukin-10/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Chronic Disease , Immune Sera/administration & dosage , Interleukin-10/antagonists & inhibitors , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukin-10/metabolism , Lymphocytic Choriomeningitis/metabolism , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Rats , Receptors, Interleukin-10/biosynthesis
5.
J Clin Invest ; 116(5): 1371-81, 2006 May.
Article in English | MEDLINE | ID: mdl-16628253

ABSTRACT

Safe induction of autoantigen-specific long-term tolerance is the "holy grail" for the treatment of autoimmune diseases. In animal models of type 1 diabetes, oral or i.n. immunization with islet antigens induces Tregs that are capable of bystander suppression. However, such interventions are only effective early in the prediabetic phase. Here, we demonstrate that a novel combination treatment with anti-CD3epsilon-specific antibody and i.n. proinsulin peptide can reverse recent-onset diabetes in 2 murine diabetes models with much higher efficacy than with monotherapy with anti-CD3 or antigen alone. In vivo, expansion of CD25(+)Foxp3(+) and insulin-specific Tregs producing IL-10, TGF-beta, and IL-4 was strongly enhanced. These cells could transfer dominant tolerance to immunocompetent recent-onset diabetic recipients and suppressed heterologous autoaggressive CD8 responses. Thus, combining a systemic immune modulator with antigen-specific Treg induction is more efficacious in reverting diabetes. Since Tregs act site-specifically, this strategy should also be expected to reduce the potential for systemic side effects.


Subject(s)
Autoantigens/therapeutic use , CD3 Complex/chemistry , Diabetes Mellitus, Type 1/prevention & control , Diabetes Mellitus, Type 1/therapy , Proinsulin/therapeutic use , T-Lymphocytes, Regulatory/metabolism , Administration, Intranasal , Animals , Autoantigens/administration & dosage , CD3 Complex/immunology , Interleukin-10/biosynthesis , Interleukin-4/biosynthesis , Mice , Mice, Transgenic , Proinsulin/administration & dosage , Remission Induction , Transforming Growth Factor beta/biosynthesis
6.
J Exp Med ; 199(8): 1153-62, 2004 Apr 19.
Article in English | MEDLINE | ID: mdl-15096540

ABSTRACT

The T-box transcription factor T-bet is known to control lineage commitment and interferon-gamma production by T helper 1 (Th1) CD4 lymphocytes. We report here that T-bet is essential for development of CD8 lymphocyte-dependent autoimmune diabetes (type 1 diabetes [T1D]) in the rat insulin promoter-lymphocytic choriomeningitis virus (LCMV) transgenic model for virally induced T1D. In the absence of T-bet, autoaggressive (anti-LCMV) CD8 lymphocytes were reduced in number and produced less IFN-gamma, but increased IL-2 compared with controls. Further analysis showed that T-bet intrinsically controls the generation, but not apoptosis, maintenance, or secondary expansion of antiviral effector/memory CD8 lymphocytes. This observation points toward a therapeutic opportunity for the treatment of T1D and other autoimmune disorders.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Transcription Factors/immunology , Animals , Autoimmunity , Chimera , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Immunologic Memory , Interferon-gamma/biosynthesis , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/pathogenicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Rats , T-Box Domain Proteins , Transcription Factors/deficiency , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...