Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Immunol ; 364: 104360, 2021 06.
Article in English | MEDLINE | ID: mdl-33866285

ABSTRACT

Ig-GAD2, an antigen-specific immune modulator, requires bone marrow (BM) cell transfer in order to restore beta (ß)-cell formation and induce recovery from established type 1 diabetes (T1D). The BM cells provide endothelial precursor cells (EPCs) that give rise to islet resident endothelial cells (ECs). This study shows that, during development of T1D, the immune attack causes collateral damage to the islet vascular network. The EPC-derived ECs repair and restore islet blood vessel integrity. In addition, ß-cell genetic tracing indicates that the newly formed ß-cells originate from residual ß-cells that escaped the immune attack and, unexpectedly, from ß-cell precursors. This indicates that the rejuvenated islet microenvironment invigorates formation of new ß-cells not only from residual ß-cells but also from precursor cells. This is twofold significant from the perspective of precursor cells as a safe reserve for restoration of ß-cell mass and its promise for therapy of T1D long after diagnosis.


Subject(s)
Bone Marrow Cells/physiology , Diabetes Mellitus, Type 1/therapy , Endothelial Progenitor Cells/physiology , Immunologic Factors/therapeutic use , Insulin-Secreting Cells/physiology , Recombinant Fusion Proteins/therapeutic use , Animals , Cell Differentiation , Cell Self Renewal , Cells, Cultured , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Glutamate Decarboxylase/genetics , Humans , Immunoglobulins/genetics , Immunologic Factors/genetics , Mice , Mice, Inbred NOD , Recombinant Fusion Proteins/genetics , Regeneration , Regional Blood Flow
2.
J Immunol ; 205(8): 2039-2045, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32917785

ABSTRACT

Early thymic progenitors (ETPs) are bone marrow-derived hematopoietic stem cells that remain multipotent and give rise to a variety of lineage-specific cells. Recently, we discovered a subset of murine ETPs that expresses the IL-4Rα/IL-13Rα1 heteroreceptor (HR) and commits only to the myeloid lineage. This is because IL-4/IL-13 signaling through the HR inhibits their T cell potential and enacts commitment of HR+ETPs to thymic resident CD11c+CD8α+ dendritic cells (DCs). In this study, we discovered that HR+-ETP-derived DCs function as APCs in the thymus and promote deletion of myelin-reactive T cells. Furthermore, this negative T cell selection function of HR+-ETP-derived DCs sustains protection against experimental allergic encephalomyelitis, a mouse model for human multiple sclerosis. These findings, while shedding light on the intricacies underlying ETP lineage commitment, reveal a novel, to our knowledge, function by which IL-4 and IL-13 cytokines condition thymic microenvironment to rheostat T cell selection and fine-tune central tolerance.


Subject(s)
Dendritic Cells/immunology , Interleukin-13/immunology , Interleukin-4/immunology , Multiple Sclerosis/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Animals , Cellular Microenvironment/genetics , Cellular Microenvironment/immunology , Dendritic Cells/pathology , Disease Models, Animal , Interleukin-13/genetics , Interleukin-4/genetics , Mice , Mice, Knockout , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , T-Lymphocytes/pathology , Thymus Gland/pathology
3.
J Immunol ; 202(11): 3173-3186, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30996000

ABSTRACT

Early life immune responses are deficient in Th1 lymphocytes that compromise neonatal vaccination. We found that IL-4 and IL-13 engage a developmentally expressed IL-4Rα/IL-13Rα1 heteroreceptor to endow IFN regulatory factor 1 (IRF-1) with apoptotic functions, which redirect murine neonatal Th1 reactivation to cell death. IL-4/IL-13-induced STAT6 phosphorylation serves to enhance IRF-1 transcription and promotes its egress from the nucleus. In the cytoplasm, IRF-1 can no longer serve as an anti-viral transcription factor but, instead, colocalizes with Bim and instigates the mitochondrial, or intrinsic, death pathway. The new pivotal function of IRF-1 in the death of neonatal Th1 cells stems from the ability of its gene to bind STAT6 for enhanced transcription and the proficiency of its protein to precipitate Bim-driven apoptosis. This cytokine-induced, IRF-1-mediated developmental death network weakens neonatal Th1 responses during early life vaccination and increases susceptibility to viral infection.


Subject(s)
Interferon Regulatory Factor-1/metabolism , Interleukin-13/metabolism , Interleukin-4/metabolism , Th1 Cells/immunology , Viral Vaccines/immunology , Virus Diseases/immunology , Animals , Animals, Newborn , Apoptosis , Bcl-2-Like Protein 11/metabolism , Disease Resistance , Humans , Immunity , Infant, Newborn , Interferon Regulatory Factor-1/genetics , Interleukin-13 Receptor alpha1 Subunit/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , STAT6 Transcription Factor/metabolism , Signal Transduction
4.
J Immunol ; 201(10): 2947-2958, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30291166

ABSTRACT

Recently we reported that IL-4 and IL-13 signaling in murine early thymic progenitors (ETPs) expressing the heteroreceptor (HR) comprising IL-4 receptor α (IL-4Rα) and IL-13 receptor α 1 (IL-13Rα1) activate STAT6 and inhibit ETP maturation potential toward T cells. In this study, we asked whether IL-4 and IL-13 signaling through the HR mobilizes other STAT molecules to shape ETP fate decision. The findings indicate that HR+ ETPs undergoing cytokine signaling display increased STAT1, but not STAT3, phosphorylation in addition to STAT6 activation. In parallel, the ETPs had a STAT1-dependent heightened expression of IRF-8, a transcription factor essential for development of CD8α+ dendritic cells (DCs). Interestingly, STAT1 phosphorylation and IRF-8 upregulation, which are independent of STAT6 activation, guided ETP maturation toward myeloid cells with a CD8α+ DC phenotype. Furthermore, these CD8α+ DCs display a thymic resident phenotype, as they did not express SIRPα, a molecule presumed to be involved in cell migration. These findings suggest that IL-4 and IL-13 cytokine-induced HR signaling provides a double-edged sword that simultaneously blocks T cell lineage potential but advances myeloid maturation that could impact T cell selection and central tolerance.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/cytology , Interleukin-13/metabolism , Interleukin-4/metabolism , Thymocytes/cytology , Animals , Central Tolerance/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Interferon Regulatory Factors/immunology , Interferon Regulatory Factors/metabolism , Interleukin-13/immunology , Interleukin-4/immunology , Mice , Mice, Knockout , STAT1 Transcription Factor/immunology , STAT1 Transcription Factor/metabolism , STAT6 Transcription Factor/immunology , STAT6 Transcription Factor/metabolism , Thymocytes/immunology , Thymocytes/metabolism
5.
Cell Immunol ; 331: 130-136, 2018 09.
Article in English | MEDLINE | ID: mdl-29929727

ABSTRACT

IL-13 receptor alpha 1 (IL-13Rα1) associates with IL-4Rα to form a functional IL-4Rα/IL-13Rα1 heteroreceptor (HR) through which both IL-4 and IL-13 signal. Recently, HR expression was associated with the development of M2 type macrophages which function as antigen presenting cells (APCs). Herein, we show that a subset of thymic resident dendritic cells (DCs) expressing high CD11b (CD11bhi) and intermediate CD11c (CD11cint) arise in HR-sufficient but not HR-deficient mice. These DCs, which originate from the bone marrow are able to take up Ag from the peritoneum, traffic through the spleen and the lymph nodes and carry it to the thymus. In addition, since the DCs are able to present Ag to T cells, express high levels of the costimulatory molecule CD24, and comprise a CD8α+ subset, it is likely that the cells contribute to T cell development and perhaps negative selection of self-reactive lymphocytes.


Subject(s)
Antigen Presentation/immunology , Cell Movement/immunology , Dendritic Cells/immunology , Interleukin-13 Receptor alpha1 Subunit/immunology , Thymus Gland/immunology , Animals , CD11c Antigen/immunology , CD11c Antigen/metabolism , CD24 Antigen/immunology , CD24 Antigen/metabolism , Dendritic Cells/metabolism , Interleukin-13 Receptor alpha1 Subunit/genetics , Interleukin-13 Receptor alpha1 Subunit/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
6.
J Immunol ; 199(8): 2767-2776, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28893952

ABSTRACT

Early thymic progenitors (ETPs) are endowed with diverse potencies and can give rise to myeloid and lymphoid lineage progenitors. How the thymic environment guides ETP commitment and maturation toward a specific lineage remains obscure. We have previously shown that ETPs expressing the heteroreceptor (HR) comprising IL-4Rα and IL-13Rα1 give rise to myeloid cells but not T cells. In this article, we show that signaling through the HR inhibits ETP maturation to the T cell lineage but enacts commitment toward the myeloid cells. Indeed, HR+ ETPs, but not HR- ETPs, exhibit activated STAT6 transcription factor, which parallels with downregulation of Notch1, a critical factor for T cell development. Meanwhile, the myeloid-specific transcription factor C/EBPα, usually under the control of Notch1, is upregulated. Furthermore, in vivo inhibition of STAT6 phosphorylation restores Notch1 expression in HR+ ETPs, which regain T lineage potential. In addition, upon stimulation with IL-4 or IL-13, HR- ETPs expressing virally transduced HR also exhibit STAT6 phosphorylation and downregulation of Notch1, leading to inhibition of lymphoid, but not myeloid, lineage potential. These observations indicate that environmental cytokines play a role in conditioning ETP lineage choice, which would impact T cell development.


Subject(s)
Interleukin-13 Receptor alpha1 Subunit/metabolism , Interleukin-13/metabolism , Interleukin-4/metabolism , Precursor Cells, T-Lymphoid/physiology , Receptors, Cell Surface/metabolism , T-Lymphocytes/physiology , Thymus Gland/immunology , Animals , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation , Cell Lineage , Cells, Cultured , Interleukin-13 Receptor alpha1 Subunit/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/physiology , Receptors, Cell Surface/genetics , STAT6 Transcription Factor/metabolism , Signal Transduction
7.
J Immunol ; 199(7): 2236-2248, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28801358

ABSTRACT

IL-4 and IL-13 have been defined as anti-inflammatory cytokines that can counter myelin-reactive T cells and modulate experimental allergic encephalomyelitis. However, it is not known whether endogenous IL-4 and IL-13 contribute to the maintenance of peripheral tolerance and whether their function is coordinated with T regulatory cells (Tregs). In this study, we used mice in which the common cytokine receptor for IL-4 and IL-13, namely the IL-4Rα/IL-13Rα1 (13R) heteroreceptor (HR), is compromised and determined whether the lack of signaling by endogenous IL-4 and IL-13 through the HR influences the function of effector Th1 and Th17 cells in a Treg-dependent fashion. The findings indicate that mice-deficient for the HR (13R-/-) are more susceptible to experimental allergic encephalomyelitis than mice sufficient for the HR (13R+/+) and develop early onset and more severe disease. Moreover, Th17 cells from 13R-/- mice had reduced ability to convert to Th1 cells and displayed reduced sensitivity to suppression by Tregs relative to Th17 effectors from 13R+/+ mice. These observations suggest that IL-4 and IL-13 likely operate through the HR and influence Th17 cells to convert to Th1 cells and to acquire increased sensitivity to suppression, leading to control of immune-mediated CNS inflammation. These previously unrecognized findings shed light on the intricacies underlying the contribution of cytokines to peripheral tolerance and control of autoimmunity.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-13 Receptor alpha1 Subunit/immunology , Receptors, Cell Surface/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Cytokines/biosynthesis , Cytokines/immunology , Disease Models, Animal , Immune Tolerance , Interleukin-13/biosynthesis , Interleukin-13/metabolism , Interleukin-13 Receptor alpha1 Subunit/deficiency , Interleukin-13 Receptor alpha1 Subunit/genetics , Interleukin-4/biosynthesis , Interleukin-4/metabolism , Mice , Mice, Inbred C57BL , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Signal Transduction , Th1 Cells/immunology
8.
J Immunol ; 199(3): 894-902, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28646042

ABSTRACT

Type 1 diabetes (T1D) manifests when the insulin-producing pancreatic ß cells are destroyed as a consequence of an inflammatory process initiated by lymphocytes of the immune system. The NOD mouse develops T1D spontaneously and serves as an animal model for human T1D. The IL-4Rα/IL-13Rα1 heteroreceptor (HR) serves both IL-4 and IL-13 cytokines, which are believed to function as anti-inflammatory cytokines in T1D. However, whether the HR provides a responsive element to environmental (i.e., physiologic) IL-4/IL-13 in the regulation of peripheral tolerance and the development of T1D has yet to be defined. In this study, NOD mice deficient for the HR have been generated by means of IL-13Rα1 gene disruption and used to determine whether such deficiency affects the development of T1D. Surprisingly, the findings indicate that NOD mice lacking the HR (13R-/-) display resistance to T1D as the rise in blood glucose level and islet inflammation were significantly delayed in these HR-deficient relative to HR-sufficient (13R+/+) mice. In fact, the frequency and spleen-to-pancreas dynamics of both Th1 and Th17 cells were affected in 13R-/- mice. This is likely due to an increase in the frequency of mTGFß+Foxp3int regulatory T cells and the persistence of CD206+ macrophages in the pancreas as both types of cells confer resistance to T1D upon transfer to 13R+/+ mice. These findings reveal new insights as to the role environmental IL-4/IL-13 and the HR play in peripheral tolerance and the development of T1D.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Interleukin-13 Receptor alpha1 Subunit/metabolism , Receptors, Cell Surface/metabolism , Adoptive Transfer , Animals , Blood Glucose , Disease Models, Animal , Insulin-Secreting Cells/immunology , Interleukin-13/immunology , Interleukin-13 Receptor alpha1 Subunit/deficiency , Interleukin-13 Receptor alpha1 Subunit/genetics , Interleukin-13 Receptor alpha1 Subunit/immunology , Interleukin-4/immunology , Lectins, C-Type/immunology , Macrophages/immunology , Mannose Receptor , Mannose-Binding Lectins/immunology , Mice , Mice, Inbred NOD , Pancreas/cytology , Pancreas/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th17 Cells/immunology
9.
J Immunol ; 197(9): 3554-3565, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27671108

ABSTRACT

To contain autoimmunity, pathogenic T cells must be eliminated or diverted from reaching the target organ. Recently, we defined a novel form of T cell tolerance whereby treatment with Ag downregulates expression of the chemokine receptor CXCR3 and prevents diabetogenic Th1 cells from reaching the pancreas, leading to suppression of type 1 diabetes (T1D). This report defines the signaling events underlying Ag-induced chemokine receptor-mediated tolerance. Specifically, we show that the mammalian target of rapamycin complex 1 (mTORC1) is a major target for induction of CXCR3 downregulation and crippling of Th1 cells. Indeed, Ag administration induces upregulation of programmed death-ligand 1 on dendritic cells in a T cell-dependent manner. In return, programmed death-ligand 1 interacts with the constitutively expressed programmed death-1 on the target T cells and stimulates docking of Src homology 2 domain-containing tyrosine phosphatase 2 phosphatase to the cytoplasmic tail of programmed death-1. Active Src homology 2 domain-containing tyrosine phosphatase 2 impairs the signaling function of the PI3K/protein kinase B (AKT) pathway, leading to functional defect of mTORC1, downregulation of CXCR3 expression, and suppression of T1D. Thus, mTORC1 component of the metabolic pathway serves as a target for chemokine receptor-mediated T cell tolerance and suppression of T1D.


Subject(s)
Multiprotein Complexes/immunology , Receptors, CXCR3/metabolism , T-Lymphocytes/immunology , TOR Serine-Threonine Kinases/immunology , Animals , Antigens/immunology , Autoimmunity , B7-H1 Antigen/metabolism , Cells, Cultured , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , Immune Tolerance , Immunomodulation , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred NOD , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...