Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Nat Immunol ; 11(11): 1057-62, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20935646

ABSTRACT

After being activated by antigen, helper T lymphocytes switch from a resting state to clonal expansion. This switch requires inactivation of the transcription factor Foxo1, a suppressor of proliferation expressed in resting helper T lymphocytes. In the early antigen-dependent phase of expansion, Foxo1 is inactivated by antigen receptor-mediated post-translational modifications. Here we show that in the late phase of expansion, Foxo1 was no longer post-translationally regulated but was inhibited post-transcriptionally by the interleukin 2 (IL-2)-induced microRNA miR-182. Specific inhibition of miR-182 in helper T lymphocytes limited their population expansion in vitro and in vivo. Our results demonstrate a central role for miR-182 in the physiological regulation of IL-2-driven helper T cell-mediated immune responses and open new therapeutic possibilities.


Subject(s)
Interleukin-2/immunology , MicroRNAs/immunology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Arthritis/immunology , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
2.
Immunity ; 28(5): 687-97, 2008 May.
Article in English | MEDLINE | ID: mdl-18468463

ABSTRACT

The dynamics of CD4(+) effector T cells (Teff cells) and CD4(+)Foxp3(+) regulatory T cells (Treg cells) during diabetes progression in nonobese diabetic mice was investigated to determine whether an imbalance of Treg cells and Teff cells contributes to the development of type 1 diabetes. Our results demonstrated a progressive decrease in the Treg cell:Teff cell ratio in inflamed islets but not in pancreatic lymph nodes. Intra-islet Treg cells expressed reduced amounts of CD25 and Bcl-2, suggesting that their decline was due to increased apoptosis. Additionally, administration of low-dose interleukin-2 (IL-2) promoted Treg cell survival and protected mice from developing diabetes. Together, these results suggest intra-islet Treg cell dysfunction secondary to defective IL-2 production is a root cause of the progressive breakdown of self-tolerance and the development of diabetes in nonobese diabetic mice.


Subject(s)
Autoimmunity , CD4-Positive T-Lymphocytes/metabolism , Diabetes Mellitus, Type 1/metabolism , Interleukin-2/metabolism , Islets of Langerhans/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Survival , Diabetes Mellitus, Type 1/immunology , Interleukin-2/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphocyte Activation , Mice , Mice, Inbred NOD , Proto-Oncogene Proteins c-bcl-2/immunology , Proto-Oncogene Proteins c-bcl-2/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism
3.
Eur J Immunol ; 45(4): 975-87, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25639669

ABSTRACT

It is believed that memory CD8(+) T cells are maintained in secondary lymphoid tissues, peripheral tissues, and BM by homeostatic proliferation. Their survival has been shown to be dependent on IL-7, but it is unclear where they acquire it. Here we show that in murine BM, memory CD8(+) T cells individually colocalize with IL-7(+) reticular stromal cells. The T cells are resting in terms of global transcription and do not express markers of activation, for example, 4-1BB (CD137), IL-2, or IFN-γ, despite the expression of CD69 on about 30% of the cells. Ninety-five percent of the memory CD8(+) T cells in BM are in G0 phase of cell cycle and do not express Ki-67. Less than 1% is in S/M/G2 of cell cycle, according to propidium iodide staining. While previous publications have estimated the extent of proliferation of CD8(+) memory T cells on the basis of BrdU incorporation, we show here that BrdU itself induces proliferation of CD8(+) memory T cells. Taken together, the present results suggest that CD8(+) memory T cells are maintained as resting cells in the BM in dedicated niches with their survival conditional on IL-7 receptor signaling.


Subject(s)
Bone Marrow Cells/cytology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Resting Phase, Cell Cycle/immunology , Stromal Cells/immunology , Animals , Antigens, CD/biosynthesis , Antigens, Differentiation, T-Lymphocyte/biosynthesis , Bone Marrow Cells/immunology , Cell Proliferation , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Interleukin-7/immunology , Ki-67 Antigen/biosynthesis , Lectins, C-Type/biosynthesis , Mice , Mice, Inbred C57BL , Transcription, Genetic , Tumor Necrosis Factor Receptor Superfamily, Member 9/biosynthesis
4.
J Immunol ; 188(3): 1064-74, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22227569

ABSTRACT

A progressive waning in Foxp3(+) regulatory T cell (Treg) functions is thought to provoke autoimmunity in the NOD model of type 1 diabetes (T1D). A deficiency in IL-2 is one of the main triggers for the defective function of Tregs in islets. Notably, abrogation of the ICOS pathway in NOD neonates or BDC2.5-NOD (BDC2.5) mice exacerbates T1D, suggesting an important role for this costimulatory pathway in tolerance to islet Ags. Thus, we hypothesize that ICOS selectively promotes Foxp3(+) Treg functions in BDC2.5 mice. We show that ICOS expression discriminates effector Foxp3(-) T cells from Foxp3(+) Tregs and specifically designates a dominant subset of intra-islet Tregs, endowed with an increased potential to expand, secrete IL-10, and mediate suppressive activity in vitro and in vivo. Consistently, Ab-mediated blockade or genetic deficiency of ICOS selectively abrogates Treg-mediated functions and T1D protection and exacerbates disease in BDC2.5 mice. Moreover, T1D progression in BDC2.5 mice is associated with a decline in ICOS expression in and expansion and suppression by intra-islet Foxp3(+) Tregs. We further show that the ICOS(+) Tregs, in contrast to their ICOS(-) counterparts, are more sensitive to IL-2, a critical signal for their survival and functional stability. Lastly, the temporal loss in ICOS(+) Tregs is readily corrected by IL-2 therapy or protective Il2 gene variation. Overall, ICOS is critical for the homeostasis and functional stability of Foxp3(+) Tregs in prediabetic islets and maintenance of T1D protection.


Subject(s)
Homeostasis/immunology , Inducible T-Cell Co-Stimulator Protein/physiology , Islets of Langerhans/immunology , T-Lymphocytes, Regulatory/physiology , Animals , Diabetes Mellitus, Type 1/prevention & control , Forkhead Transcription Factors , Interleukin-2/therapeutic use , Mice , Mice, Inbred NOD , Prediabetic State/immunology
5.
J Immunol ; 186(12): 6788-97, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21576508

ABSTRACT

Natural FOXP3(+)CD4(+)CD25(High) regulatory T cells are critical in immunological self-tolerance. Their characterization in humans is hindered by the failure to discriminate these cells from activated effector T cells in inflammation. To explore the relationship between FOXP3 expression and regulatory function at the clonal level, we used a single-cell cloning strategy of CD25-expressing CD4(+) T cell subsets from healthy human donors. Our approach unveils a functional heterogeneity nested within CD4(+)CD25(High)FOXP3(+) T cells, and typically not revealed by conventional bulk assays. Whereas most cells display the canonical regulatory T (T(reg)) cell characteristics, a significant proportion of FOXP3(+) T cells is compromised in its suppressive function, despite the maintenance of other phenotypic and functional regulatory T hallmark features. In addition, these nonsuppressive FOXP3(+) T cells preferentially emerge from the CD45RO(+) memory pool, and arise as a consequence of a rapid downregulation of FOXP3 expression upon T cell reactivation. Surprisingly, these dysfunctional T(reg) cells with unstable FOXP3 expression do not manifest overt plasticity in terms of inflammatory cytokine secretion. These results open a path to an extensive study of the functional heterogeneity of CD4(+)CD25(High)FOXP3(+) T(reg) cells and warrant caution in the sole use of FOXP3 as a clinical marker for monitoring of immune regulation in humans.


Subject(s)
Forkhead Transcription Factors/analysis , T-Lymphocytes, Regulatory/immunology , Antigens, CD/analysis , Antigens, CD/immunology , Biomarkers/analysis , Clone Cells/immunology , Forkhead Transcription Factors/genetics , Humans , Immunologic Memory/immunology , Inflammation Mediators/analysis , Lymphocyte Activation/immunology , Self Tolerance/immunology , T-Lymphocytes, Regulatory/cytology
6.
J Immunol ; 181(9): 6283-92, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18941219

ABSTRACT

Type I diabetes (T1D) susceptibility is inherited through multiple insulin-dependent diabetes (Idd) genes. NOD.B6 Idd3 congenic mice, introgressed with an Idd3 allele from T1D-resistant C57BL/6 mice (Idd3(B6)), show a marked resistance to T1D compared with control NOD mice. The protective function of the Idd3 locus is confined to the Il2 gene, whose expression is critical for naturally occurring CD4(+)Foxp3(+) regulatory T (nT(reg)) cell development and function. In this study, we asked whether Idd3(B6) protective alleles in the NOD mouse model confer T1D resistance by promoting the cellular frequency, function, or homeostasis of nT(reg) cells in vivo. We show that resistance to T1D in NOD.B6 Idd3 congenic mice correlates with increased levels of IL-2 mRNA and protein production in Ag-activated diabetogenic CD4(+) T cells. We also observe that protective IL2 allelic variants (Idd3(B6) resistance allele) also favor the expansion and suppressive functions of CD4(+)Foxp3(+) nT(reg) cells in vitro, as well as restrain the proliferation, IL-17 production, and pathogenicity of diabetogenic CD4(+) T cells in vivo more efficiently than control do nT(reg) cells. Lastly, the resistance to T1D in Idd3 congenic mice does not correlate with an augmented systemic frequency of CD4(+)Foxp3(+) nT(reg) cells but more so with the ability of protective IL2 allelic variants to promote the expansion of CD4(+)Foxp3(+) nT(reg) cells directly in the target organ undergoing autoimmune attack. Thus, protective, IL2 allelic variants impinge the development of organ-specific autoimmunity by bolstering the IL-2 producing capacity of self-reactive CD4(+) T cells and, in turn, favor the function and homeostasis of CD4(+)Foxp3(+) nT(reg) cells in vivo.


Subject(s)
Alleles , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Forkhead Transcription Factors/biosynthesis , Genetic Variation/physiology , Interleukin-2/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Cells, Cultured , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Disease Progression , Genetic Predisposition to Disease , Homeostasis/genetics , Homeostasis/immunology , Immunity, Cellular/genetics , Immunity, Innate/genetics , Interleukin-2/metabolism , Interleukin-2/physiology , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Pancreas/cytology , Pancreas/immunology , Pancreas/metabolism , T-Lymphocytes, Regulatory/pathology
7.
Diabetes Metab Res Rev ; 25(3): 208-18, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19214972

ABSTRACT

In recent years, there has been a revival of the concept of CD4(+) regulatory T (T(reg)) cells as being a central control point in various immune responses, including autoimmune responses and immunity to transplants, allergens, tumours and infectious microbes. The current literature suggests that T(reg) cells are diverse in their phenotype and mechanism(s) of action, and as such, may constitute a myriad of naturally occurring and induced T cell precursors with variable degrees of regulatory potential. In this review, we summarize research from various laboratories, including our own, showing that CD4(+)Foxp3(+) T(reg) cells are critical in the control of type 1 diabetes (T1D) in mouse models and humans. In this review, we also discuss cellular and molecular determinants that impact CD4(+)Foxp3(+) T(reg) cell development and function and consequential resistance to organ-specific autoimmune disease. Recent advances in the use of CD4(+)Foxp3(+) T(reg) cellular therapy to promote immunological tolerance in the absence of long-term generalized immunosuppression are also presented.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Forkhead Transcription Factors/physiology , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmune Diseases/immunology , CD4 Antigens/immunology , Dendritic Cells/physiology , Humans , Interleukin-2/physiology , Mice , Mice, Inbred NOD , Pancreas/immunology
8.
Ann N Y Acad Sci ; 1247: 46-55, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22236474

ABSTRACT

The Forkhead box O (FoxO) family of transcription factors is important for the maintenance of immunological homeostasis and tolerance by controlling the development and function of B and T lymphocytes. Because dysregulation in FoxO activity can result in chronic inflammation and autoimmunity, the transcriptional activity of FoxO proteins is tightly controlled and generally dependent on complex posttranslational modifications that lead either to their nuclear entry and subsequent activation or, alternatively, to their nuclear export. The phosphatidylinositol 3-kinase (PI3K)-protein kinase B (PKB/Akt) axis represents the major pathway phosphorylating and thereby inactivating FoxO proteins. However, recent results have revealed an additional posttranscriptional mechanism of FoxO inactivation by microRNAs. The discovery of this molecular pathway may provide a new therapeutic avenue for the modulation of FoxO activity in immune-mediated diseases using either microRNA targeting antagomirs or synthetic microRNA mimics, a topic that is addressed in this review.


Subject(s)
Forkhead Transcription Factors/genetics , Lymphocytes/metabolism , MicroRNAs/metabolism , Signal Transduction , Adaptive Immunity/genetics , Animals , Forkhead Transcription Factors/metabolism , Humans , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Processing, Post-Transcriptional
9.
Autoimmunity ; 44(5): 406-14, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21244339

ABSTRACT

Il2 allelic variation in non-obese diabetic mice imparts marked resistance to type 1 diabetes. IL-2 is pivotal for the fitness and homeostasis of Foxp3(+) regulatory T (T(reg)) cells, and the Idd3(B6) locus augments IL-2 production by effector T cells, which in turn enhances the potency of T(reg) cell functions. Given the important role dendritic cells (DCs) play in T(reg) cell-mediated tolerance induction, we hypothesized that DCs from Idd3(B6) congenic mice contribute to increased T(reg) cell activity. Here, we observed that CD11c(+) DCs, harboring protective Idd3(B6) genes, are endowed with the capacity to secrete IL-2, enabling them to preferentially promote T(reg) cell functions in vitro and in vivo. Our results show that Il2 gene variation may imprint DCs to favor T-cell regulation of autoimmunity.


Subject(s)
Autoimmunity/immunology , Dendritic Cells/immunology , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Interleukin-2/biosynthesis , Interleukin-2/metabolism , T-Lymphocytes, Regulatory/immunology , Alleles , Animals , Autoimmunity/genetics , Dendritic Cells/metabolism , Female , Genetic Variation/genetics , Genomic Imprinting/genetics , Interleukin-2/genetics , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Inbred NOD , Pancreas/immunology , Pancreas/metabolism , Phenotype , T-Lymphocytes, Regulatory/cytology
10.
Curr Opin Immunol ; 20(6): 655-62, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18926906

ABSTRACT

The immune system requires a homeostatic equilibrium between the mechanisms that assure self-tolerance, those that control the capacity to mount life-long immunity to pathogenic microbes, and those that attenuate effector mechanisms from inducing immune pathology [Sakaguchi S, Yamaguchi T, Nomura T, Ono M: Regulatory T cells and immune tolerance. Cell 2008, 133 (5):775-87; Piccirillo CA, Thornton AM: Cornerstone of peripheral tolerance: naturally occurring CD4+CD25+regulatory T cells. Trends Immunol 2004, 25:374-80]. In the past decade, an overwhelming body of literature showed that CD4+Foxp3+ regulatory T (Treg) cells are a dominant mechanism regulating the decision fate of these different immunological outcomes. Indeed, CD4+Foxp3+ Treg cells develop largely in the thymus but can be induced in the periphery throughout the course of immune responses [Sakaguchi S, Yamaguchi T, Nomura T, Ono M: Regulatory T cells and immune tolerance. Cell 2008, 133 (5):775-87; Piccirillo CA, Thornton AM: Cornerstone of peripheral tolerance: naturally occurring CD4+CD25+regulatory T cells. Trends Immunol 2004, 25:374-80]. Treg cells have emerged as a central control point in the regulation of autoimmune responses. Despite progress made in various in vitro and in vivo models, much uncertainty exists over their mechanism of action in vivo. Here, we summarize research characterizing the functional dynamics of CD4+Foxp3+ Treg cells in the control of autoimmunity in rodents and humans.


Subject(s)
Autoimmune Diseases/immunology , Autoimmunity/immunology , Cytokines/immunology , Self Tolerance/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmune Diseases/metabolism , CD4 Antigens/immunology , Cytokines/metabolism , Forkhead Transcription Factors/immunology , Humans , Inflammation/immunology , Inflammation/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism
11.
Diabetes ; 57(1): 113-23, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17928397

ABSTRACT

OBJECTIVE: In this study, we asked whether a possible quantitative or qualitative deficiency in naturally occurring Foxp3(+)CD4(+) regulatory T-cells (nT(reg)), which display potent inhibitory effects on T-cell functions in vitro and in vivo, may predispose to the development of type 1 diabetes. RESEARCH DESIGN AND METHODS: We assessed the frequency and function of Foxp3(+) nT(reg) cells in primary and secondary lymphoid tissues in the NOD animal model of type 1 diabetes. RESULTS: We show that the cellular frequency of Foxp3(+) nT(reg) cells in primary and secondary lymphoid tissues is stable and does not decline relative to type 1 diabetes-resistant mice. We show that thymic and peripheral CD4(+)CD25(+) T-cells are fully functional in vivo. We also examined the functional impact of CD4(+)Foxp3(+) nT(reg) cells on the development of autoimmune diabetes, and we demonstrate that nT(reg) cells do not affect the initial priming or expansion of antigen-specific diabetogenic T-cells but impact their differentiation in pancreatic lymph nodes. Moreover, CD4(+)Foxp3(+) nT(reg) cells also regulate later events of diabetogenesis by preferentially localizing in the pancreatic environment where they suppress the accumulation and function of effector T-cells. Finally, we show that the nT(reg) cell functional potency and intra-pancreatic proliferative potential declines with age, in turn augmenting diabetogenic responses and disease susceptibility. CONCLUSIONS: This study demonstrates that Foxp3-expressing nT(reg) cells in NOD mice regulate diabetogenesis, but temporal alterations in nT(reg) cell function promote immune dysregulation and the onset of spontaneous autoimmunity.


Subject(s)
Diabetes Mellitus, Type 1/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Division , Cytokines/immunology , Cytokines/physiology , Diabetes Mellitus, Type 1/pathology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/physiology , Islets of Langerhans , Ki-67 Antigen/physiology , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Pancreas/pathology , Phenotype , T-Lymphocyte Subsets/immunology
SELECTION OF CITATIONS
SEARCH DETAIL