Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Nat Immunol ; 17(5): 583-92, 2016 May.
Article in English | MEDLINE | ID: mdl-26998763

ABSTRACT

Interleukin 1ß (IL-1ß) is critical for the in vivo survival, expansion and effector function of IL-17-producing helper T (T(H)17) cells during autoimmune responses, including experimental autoimmune encephalomyelitis (EAE). However, the spatiotemporal role and cellular source of IL-1ß during EAE pathogenesis are poorly defined. In the present study, we uncovered a T cell-intrinsic inflammasome that drives IL-1ß production during T(H)17-mediated EAE pathogenesis. Activation of T cell antigen receptors induced expression of pro-IL-1ß, whereas ATP stimulation triggered T cell production of IL-1ß via ASC-NLRP3-dependent caspase-8 activation. IL-1R was detected on T(H)17 cells but not on type 1 helper T (T(H)1) cells, and ATP-treated T(H)17 cells showed enhanced survival compared with ATP-treated T(H)1 cells, suggesting autocrine action of T(H)17-derived IL-1ß. Together these data reveal a critical role for IL-1ß produced by a T(H)17 cell-intrinsic ASC-NLRP3-caspase-8 inflammasome during inflammation of the central nervous system.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , T-Lymphocytes/immunology , Th17 Cells/immunology , Adenosine Triphosphate/pharmacology , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Carrier Proteins/genetics , Carrier Proteins/immunology , Carrier Proteins/metabolism , Caspase 8/genetics , Caspase 8/immunology , Caspase 8/metabolism , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Flow Cytometry , Gene Expression/immunology , Immunoblotting , Inflammasomes/genetics , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Th17 Cells/drug effects , Th17 Cells/metabolism
2.
J Autoimmun ; 146: 103228, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38642507

ABSTRACT

CD6 is a glycoprotein expressed on CD4 and CD8 T cells involved in immunoregulation. CD318 has been identified as a CD6 ligand. The role of CD318 in T cell immunity is restricted as it has only been investigated in a few mice autoimmune models but not in human diseases. CD318 expression was thought to be limited to mesenchymal-epithelial cells and, therefore, contribute to CD6-mediated T cell activation in the CD318-expressing tissue rather than through interaction with antigen-presenting cells. Here, we report CD318 expression in a subpopulation of CD318+ myeloid dendritic (mDC), whereas the other peripheral blood populations were CD318 negative. However, CD318 can be induced by activation: a subset of monocytes treated with LPS and IFNγ and in vitro monocyte derived DCs were CD318+. We also showed that recombinant CD318 inhibited T cell function. Strikingly, CD318+ DCs suppressed the proliferation of autoreactive T cells specific for GAD65, a well-known targeted self-antigen in Type 1 Diabetes (T1D). Our study provides new insight into the role of the CD318/CD6 axis in the immunopathogenesis of inflammation, suggesting a novel immunoregulatory role of CD318 in T cell-mediated autoimmune diseases and identifying a potential novel immune checkpoint inhibitor as a target for intervention in T1D which is an unmet therapeutic need.


Subject(s)
Antigens, CD , Autoantigens , Dendritic Cells , Diabetes Mellitus, Type 1 , Islets of Langerhans , Lymphocyte Activation , Humans , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Lymphocyte Activation/immunology , Autoantigens/immunology , Antigens, CD/metabolism , Antigens, CD/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Antigens, Differentiation, T-Lymphocyte/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Cells, Cultured , Glutamate Decarboxylase
3.
Immunity ; 36(5): 821-33, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22608496

ABSTRACT

Interleukin-25 (IL-25 or IL-17E), a member of the structurally related IL-17 family, functions as an important mediator of T helper 2 cell-type (type 2) responses. We examined the cell type-specific role of IL-25-induced Act1-mediated signaling in protective immunity against helminth infection. Targeted Act1 deficiency in epithelial cells resulted in a marked delay in worm expulsion and abolished the expansion of the Lin(-)c-Kit(+) innate cell population in the mesenteric lymph node, lung, and liver. Th2 cell-inducing cytokine (IL-25 and IL-33) expression were reduced in the intestinal epithelial cells from the infected and IL-25-injected epithelial-specific Act1-deficient mice. Adoptive transfer of Lin(-)c-Kit(+) cells or combined injection of IL-25 and IL-33 restored the type 2 responses in these mice. Taken together, these results suggest that epithelial-specific Act1 mediates the expansion of the Lin(-)c-Kit(+) innate cell population through the positive-feedback loop of IL-25, initiating the type 2 immunity against helminth infection.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Epithelial Cells/immunology , Helminthiasis/immunology , Helminths/immunology , Interleukins/immunology , Th2 Cells/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Lineage , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/metabolism , Helminthiasis/metabolism , Helminths/metabolism , Immunity, Innate/immunology , Interleukins/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Liver/cytology , Liver/immunology , Liver/metabolism , Lung/cytology , Lung/immunology , Lung/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Th2 Cells/metabolism
4.
Immunity ; 32(1): 54-66, 2010 Jan 29.
Article in English | MEDLINE | ID: mdl-20060329

ABSTRACT

Interleukin-1 (IL-1)-mediated signaling in T cells is essential for T helper 17 (Th17) cell differentiation. We showed here that SIGIRR, a negative regulator of IL-1 receptor and Toll-like receptor signaling, was induced during Th17 cell lineage commitment and governed Th17 cell differentiation and expansion through its inhibitory effects on IL-1 signaling. The absence of SIGIRR in T cells resulted in increased Th17 cell polarization in vivo upon myelin oligodendrocyte glycoprotein (MOG(35-55)) peptide immunization. Recombinant IL-1 promoted a marked increase in the proliferation of SIGIRR-deficient T cells under an in vitro Th17 cell-polarization condition. Importantly, we detected increased IL-1-induced phosphorylation of JNK and mTOR kinase in SIGIRR-deficient Th17 cells compared to wild-type Th17 cells. IL-1-induced proliferation was abolished in mTOR-deficient Th17 cells, indicating the essential role of mTOR activation. Our results demonstrate an important mechanism by which SIGIRR controls Th17 cell expansion and effector function through the IL-1-induced mTOR signaling pathway.


Subject(s)
Cell Differentiation/immunology , Interleukin-17/immunology , Intracellular Signaling Peptides and Proteins/immunology , Protein Serine-Threonine Kinases/immunology , Receptors, Interleukin-1/immunology , T-Lymphocytes, Helper-Inducer/cytology , Animals , Cell Lineage/immunology , Cell Proliferation , Cell Separation , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Enzyme Activation/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunoblotting , Immunoprecipitation , Interleukin-17/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/metabolism , Receptors, Interleukin-1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , TOR Serine-Threonine Kinases , Transfection
5.
J Immunol ; 198(2): 908-915, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27927968

ABSTRACT

γδ T lymphocytes, dominant T cell subsets in the intestine, mediate both regulatory and pathogenic roles, yet the mechanisms underlying such opposing effects remain unclear. In this study, we identified a unique γδ T cell subset that coexpresses high levels of gut-homing integrins, CD103 and α4ß7. They were exclusively found in the mesenteric lymph node after T cell-mediated colitis induction, and their appearance preceded the inflammation. Adoptive transfer of the CD103+α4ß7high subsets enhanced Th1/Th17 T cell generation and accumulation in the intestine, and the disease severity. The level of generation correlated with the disease severity. Moreover, these cells were also found to be elevated in a spontaneous mouse model of ileitis. Based on the procolitogenic function, we referred to this subset as "inflammatory" γδ T cells. Targeting inflammatory γδ T cells may open a novel strategy to treat inflammatory diseases where γδ T cells play a pathogenic role including inflammatory bowel disease.


Subject(s)
Antigens, CD/immunology , Inflammatory Bowel Diseases/immunology , Integrin alpha Chains/immunology , Integrins/immunology , T-Lymphocyte Subsets/immunology , Adoptive Transfer , Animals , Disease Models, Animal , Flow Cytometry , Gene Expression Profiling , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Lymphocyte Homing/immunology , Transcriptome
6.
J Immunol ; 192(6): 2537-41, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24523506

ABSTRACT

Naive CD4 T cells transferred into lymphopenic mice undergo spontaneous proliferation and induce chronic inflammation in the intestine. Cellular mechanisms regulating the proliferative and inflammatory processes are not fully understood. In this study, we report that IFN-γ signaling in host cells plays a major role in limiting both T cell expansion and T cell-induced intestinal inflammation. However, the role of IFN-γ appears to differ depending on the target cells. IFN-γ signaling in dendritic cells controls T cell expansion, whereas IFN-γ signaling in neutrophils seems to regulate both T cell expansion and inflammation. IFN-γ signaling in nonhematopoietic cells may control inflammation. Therefore, our results suggest novel immunoregulatory functions for IFN-γ to orchestrate colitogenic T cell responses through its distinct action on different non-T cell target cells.


Subject(s)
Inflammation/immunology , Intestines/immunology , Receptors, Interferon/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , Cell Proliferation , Dendritic Cells/immunology , Dendritic Cells/metabolism , Flow Cytometry , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Immunohistochemistry , Inflammation/genetics , Inflammation/metabolism , Intestinal Mucosa/metabolism , Intestines/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Neutrophils/metabolism , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Signal Transduction/genetics , T-Lymphocytes/metabolism , Interferon gamma Receptor
7.
Proc Natl Acad Sci U S A ; 109(31): 12698-703, 2012 Jul 31.
Article in English | MEDLINE | ID: mdl-22802622

ABSTRACT

Here we report a unique role for MHC II-peptide complexes in controlling immune responses of naïve CD8 T cells. Compared with CD8 T cells from WT mice, CD8 T cells isolated from MHC II(-/-) mice hyperproliferated under lymphopenic conditions, differentiated into effector cells producing proinflammatory cytokines, and mediated more severe tissue inflammation. The elevated responses of MHC II(-/-) CD8 T cells were due to the absence of MHC II, but not CD4, T cells. The hyperreactivity appeared to be a feature of mature T cells, given its absence in CD8 single positive thymocytes derived from MHC II(-/-) mice. Expression of the MHC II ligand LAG3 was markedly enhanced during in vivo activation of MHC II(-/-) CD8 T cells, and blockade of MHC II-LAG3 interactions further enhanced T-cell expansion. Importantly, CD8 T cells isolated from H-2M(-/-) mice expressing WT levels of MHC II also displayed hyperresponsiveness similar to that of MHC II(-/-) CD8 T cells, suggesting that peptides presented on MHC II are involved in the control of CD8 T-cell responses. Our results uncover a previously undefined MHC II-dependent regulation that tunes CD8 T-cell reactivity and may have implications for an improved understanding of CD8 T-cell homeostasis and functions.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Histocompatibility Antigens Class II/immunology , Immunity, Cellular , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Cell Differentiation/genetics , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Histocompatibility Antigens Class II/genetics , Mice , Mice, Knockout , Lymphocyte Activation Gene 3 Protein
8.
Immunol Cell Biol ; 92(1): 90-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24189163

ABSTRACT

Disturbance of T-cell homeostasis could lead to intestinal inflammation. Naive CD4 T cells undergoing spontaneous proliferation, a robust proliferative response that occurs under severe lymphopenic conditions, differentiate into effector cells producing Th1- and/or Th17-type cytokines and induce a chronic inflammation in the intestine that resembles human inflammatory bowel disease. In this study, we investigated the key properties of CD4 T cells necessary to induce experimental colitis. α4ß7 upregulation was primarily induced by mesenteric lymph node (mLN) resident CD11b(+) dendritic cell subsets via transforming growth factor beta (TGFß)/retinoic acid-dependent mechanism. Interestingly, α4ß7 expression was essential but not sufficient to induce inflammation. In addition to gut-homing specificity, expression of gut Ag specificity was also crucial. T-cell acquisition of the specificity was dramatically enhanced by the presence of γδ T cells, a population previously shown to exacerbate T-cell-mediated colitis. Importantly, interleukin (IL)-23-mediated γδ T cell stimulation was necessary to enhance colitogenicity but not gut antigen reactivity of proliferating CD4 T cells. These findings demonstrate that T-cell colitogenicity is achieved through multiple processes, offering a therapeutic rationale by intervening these pathways.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Colitis/immunology , Gastrointestinal Tract/immunology , Integrins/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Th17 Cells/immunology , Animals , Antineoplastic Agents/pharmacology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Colitis/metabolism , Colitis/pathology , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/pathology , Genes, T-Cell Receptor beta/physiology , Homeodomain Proteins/physiology , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Interleukin-16/physiology , Interleukin-23 Subunit p19/physiology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Mesenteric Veins/immunology , Mesenteric Veins/metabolism , Mesenteric Veins/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Th17 Cells/cytology , Th17 Cells/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tretinoin/pharmacology
9.
J Immunol ; 188(1): 230-7, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22116827

ABSTRACT

Naive T cells undergo robust proliferation in lymphopenic conditions, whereas they remain quiescent in steady-state conditions. However, a mechanism by which naive T cells are kept from proliferating under steady-state conditions remains unclear. In this study, we report that memory CD4 T cells are able to limit naive T cell proliferation within lymphopenic hosts by modulating stimulatory functions of dendritic cells (DC). The inhibition was mediated by IL-27, which was primarily expressed in CD8(+) DC subsets as the result of memory CD4 T cell-DC interaction. IL-27 appeared to be the major mediator of inhibition, as naive T cells deficient in IL-27R were resistant to memory CD4 T cell-mediated inhibition. Finally, IL-27-mediated regulation of T cell proliferation was also observed in steady-state conditions as well as during Ag-mediated immune responses. We propose a new model for maintaining peripheral T cell homeostasis via memory CD4 T cells and CD8(+) DC-derived IL-27 in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8 Antigens , Cell Proliferation , Dendritic Cells/immunology , Gene Expression Regulation/immunology , Immunologic Memory , Interleukins/immunology , Models, Immunological , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Communication/genetics , Cell Communication/immunology , Dendritic Cells/metabolism , Gene Expression Regulation/genetics , Homeostasis/genetics , Homeostasis/immunology , Interleukins/biosynthesis , Interleukins/genetics , Mice , Mice, Knockout
10.
Cells ; 13(11)2024 May 22.
Article in English | MEDLINE | ID: mdl-38891024

ABSTRACT

BACH2 (BTB Domain and CNC Homolog 2) is a transcription factor that serves as a central regulator of immune cell differentiation and function, particularly in T and B lymphocytes. A picture is emerging that BACH2 may function as a master regulator of cell fate that is exquisitely sensitive to cell activation status. In particular, BACH2 plays a key role in stabilizing the phenotype and suppressive function of transforming growth factor-beta (TGF-ß)-derived human forkhead box protein P3 (FOXP3)+ inducible regulatory T cells (iTregs), a cell type that holds great clinical potential as a cell therapeutic for diverse inflammatory conditions. As such, BACH2 potentially could be targeted to overcome the instability of the iTreg phenotype and suppressive function that has hampered their clinical application. In this review, we focus on the role of BACH2 in T cell fate and iTreg function and stability. We suggest approaches to modulate BACH2 function that may lead to more stable and efficacious Treg cell therapies.


Subject(s)
Basic-Leucine Zipper Transcription Factors , T-Lymphocytes, Regulatory , Humans , T-Lymphocytes, Regulatory/immunology , Basic-Leucine Zipper Transcription Factors/metabolism , Animals , Forkhead Transcription Factors/metabolism , Cell- and Tissue-Based Therapy/methods , Cell Differentiation
11.
J Immunol ; 186(8): 4546-50, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21402889

ABSTRACT

Th 17 cells have been implicated in the pathogenesis of colitis; however, a cellular mechanism by which colitogenic Th17 immunity arises in vivo remains unclear. In this study, we report that a subset of IL-17(+) γδ T cells plays a crucial role in enhancing in vivo Th17 differentiation and T cell-mediated colitis. TCRß(-/-) mice were highly susceptible to T cell-mediated colitis, whereas TCRßδ(-/-) mice were resistant to the disease. Importantly, cotransfer of IL-17(+) but not of IL-17(-) γδ T cells with CD4 T cells was sufficient to enhance Th17 differentiation and induce full-blown colitis in TCRßδ(-/-) recipients. Collectively, our results provide a novel function of IL-17(+) γδ T cell subsets in supporting in vivo Th17 differentiation and possibly in fostering the development of intestinal inflammation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Colitis/immunology , Interleukin-17/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Colitis/genetics , Colitis/metabolism , Flow Cytometry , Genetic Predisposition to Disease/genetics , Immunity, Innate/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-17/genetics , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation , Th17 Cells/immunology , Th17 Cells/metabolism
12.
Sci Adv ; 9(44): eadf5238, 2023 11 03.
Article in English | MEDLINE | ID: mdl-37910614

ABSTRACT

Treatment for type 1 diabetes (T1D) requires stimulation of functional ß cell regeneration and survival under stress. Previously, we showed that inhibition of the RANKL/RANK [receptor activator of nuclear factor kappa Β (NF-κB) ligand] pathway, by osteoprotegerin and the anti-osteoporotic drug denosumab, induces rodent and human ß cell proliferation. We demonstrate that the RANK pathway mediates cytokine-induced rodent and human ß cell death through RANK-TRAF6 interaction and induction of NF-κB activation. Osteoprotegerin and denosumab protected ß cells against this cytotoxicity. In human immune cells, osteoprotegerin and denosumab reduce proinflammatory cytokines in activated T-cells by inhibiting RANKL-induced activation of monocytes. In vivo, osteoprotegerin reversed recent-onset T1D in nonobese diabetic/Ltj mice, reduced insulitis, improved glucose homeostasis, and increased plasma insulin, ß cell proliferation, and mass in these mice. Serum from T1D subjects induced human ß cell death and dysfunction, but not α cell death. Osteoprotegerin and denosumab reduced T1D serum-induced ß cell cytotoxicity and dysfunction. Inhibiting RANKL/RANK could have therapeutic potential.


Subject(s)
Diabetes Mellitus, Type 1 , Osteoprotegerin , Humans , Mice , Animals , Osteoprotegerin/metabolism , Cytokines , Diabetes Mellitus, Type 1/drug therapy , Receptor Activator of Nuclear Factor-kappa B/metabolism , Denosumab/pharmacology , NF-kappa B/metabolism , Rodentia/metabolism , RANK Ligand/metabolism , Cell Death
13.
Immunol Cell Biol ; 90(4): 396-403, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21647171

ABSTRACT

A proportional balance between αß and γδ T-cell subsets in the periphery is exceedingly well maintained by a homeostatic mechanism. However, a cellular mechanism underlying the regulation remains undefined. We recently reported that a subset of developing γδ T cells spontaneously acquires interleukin (IL)-17-producing capacity even within naive animals through a transforming growth factor (TGF)ß1-dependent mechanism, thus considered 'innate' IL-17-producing cells. Here, we report that γδ T cells generated within αß T cell (or CD4 T cell)-deficient environments displayed altered cytokine profiles; particularly, 'innate' IL-17 expression was significantly impaired compared with those in wild-type mice. Impaired IL-17 production in γδ T cells was directly related to CD4 T-cell deficiency, because depletion of CD4 T cells in wild-type mice diminished and adoptive CD4 T-cell transfer into T-cell receptor ß-/- mice restored IL-17 expression in γδ T cells. CD4 T cell-mediated IL-17 expression required TGFß1. Moreover, Th17 but not Th1 or Th2 effector CD4 T cells were highly efficient in enhancing γδ T-cell IL-17 expression. Taken together, our results highlight a novel CD4 T cell-dependent mechanism that shapes the generation of IL-17+ γδ T cells in naive settings.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Interleukin-17/biosynthesis , T-Lymphocyte Subsets/metabolism , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , Homeostasis , Immunity, Innate , Mice , Receptors, Antigen, T-Cell, gamma-delta , Transforming Growth Factor beta
14.
Cell Immunol ; 272(2): 117-23, 2012.
Article in English | MEDLINE | ID: mdl-22169530

ABSTRACT

Within lymphopenic recipients, naïve T cells undergo proliferation that is induced by homeostatic mechanisms. Earlier studies have demonstrated that commensal antigens play a key role in inducing the proliferation. However, a relative contribution of endogenous self antigens in this process has not been formally investigated. In this study, we utilized a pharmacologic inhibitor that blocks T cell egress from the lymphoid tissues, antibiotics, and germ-free animals to examine the role of commensal and self antigens. The results suggest that T cell proliferation under lymphopenic conditions is a heterogeneous process triggered by both exogenous commensal and endogenous self antigens.


Subject(s)
Autoantigens/immunology , Lymphopenia/immunology , T-Lymphocytes/immunology , Animals , Cell Proliferation/drug effects , Fingolimod Hydrochloride , Homeostasis/drug effects , Homeostasis/immunology , Immunologic Memory/drug effects , Immunologic Memory/immunology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Propylene Glycols/pharmacology , Receptors, Antigen, T-Cell/immunology , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Spleen/drug effects , Spleen/immunology , T-Lymphocytes/drug effects
15.
J Immunol ; 184(4): 1675-9, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20061408

ABSTRACT

In naive animals, gammadelta T cells are innate sources of IL-17, a potent proinflammatory cytokine mediating bacterial clearance as well as autoimmunity. However, mechanisms underlying the generation of these cells in vivo remain unclear. In this study, we show that TGF-beta1 plays a key role in the generation of IL-17(+) gammadelta T cells and that it mainly occurs in the thymus particularly during the postnatal period. Interestingly, IL-17(+) gammadelta TCR(+) thymocytes were mainly CD44(high)CD25(low) cells, which seem to derive from double-negative 4 gammadelta TCR(+) cells that acquired CD44 and IL-17 expression. Our findings identify a novel developmental pathway during which IL-17-competent gammadelta T cells arise in the thymus by a TGF-beta1-dependent mechanism.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Interleukin-17/biosynthesis , Receptors, Antigen, T-Cell, gamma-delta/biosynthesis , Thymus Gland/immunology , Thymus Gland/metabolism , Transforming Growth Factor beta1/physiology , Animals , Animals, Newborn , Cell Differentiation/genetics , Interleukin-17/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Transforming Growth Factor beta/biosynthesis , Receptors, Transforming Growth Factor beta/genetics , Thymus Gland/cytology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
16.
Proc Natl Acad Sci U S A ; 106(48): 20394-8, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19920180

ABSTRACT

T cells transferred into severe lymphopenic hosts undergo rapid proliferation known as "endogenous proliferation" that are distinct from conventional homeostatic proliferation. Unlike homeostatic proliferation, cytokines, such as IL-7 are dispensable, yet TCR:MHC interaction is essential for this process to occur. However, cell types inducing the proliferation have not formally been addressed. In this study, we report that CD11c+ conventional DCs play irreplaceable roles in inducing endogenous proliferation of both naive and memory phenotype CD4 T cells via TCR-MHC II interaction. By contrast, CD8 T-cell endogenous proliferation was independent of MHC I or CD11c+ DCs. Interestingly, MHC II was necessary to support naive CD8 T-cell proliferation within MHC I-deficient hosts. Depletion of both B cells and DCs was sufficient to abrogate the proliferation of naive but not of memory CD8 T cells. These results suggest that depending on the T-cell lineages, as well as the differentiation status, different mechanisms control endogenous proliferation, revealing in vivo complexity of T-cell proliferation under lymphopenic conditions.


Subject(s)
Cell Proliferation , Dendritic Cells/immunology , T-Lymphocyte Subsets/immunology , Adoptive Transfer , Animals , B-Lymphocytes/immunology , CD11c Antigen/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/metabolism , Flow Cytometry , Genes, MHC Class II/genetics , Genes, T-Cell Receptor beta/genetics , Mice , Mice, Knockout
17.
Blood ; 113(25): 6361-71, 2009 Jun 18.
Article in English | MEDLINE | ID: mdl-19380870

ABSTRACT

Homeostatic mechanism by which peripheral T-cell subsets are maintained in vivo remains largely unknown. Using a T-cell proliferation model under lymphopenic settings, we now demonstrate that gammadelta T cells limit CD8 T-cell expansion but not the initial proliferation after transfer into lymphopenic recipients. Interleukin-15 (IL-15) produced by and trans-presented on the membrane of the CD11c(+) dendritic cells (DCs) is the key factor that mediates homeostatic competition between CD8 and gammadelta T cells, revealing previously unrecognized IL-15-dependent homeostatic mechanisms between different T-cell subsets in vivo.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Interleukin-15/physiology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , Adoptive Transfer , Animals , CD11c Antigen/analysis , Cell Division , Genes, RAG-1 , Homeostasis/immunology , Interleukin-15/deficiency , Interleukin-15/genetics , Interleukin-15/immunology , Interleukin-15/pharmacology , Lymphocyte Activation , Lymphocyte Depletion , Lymphopenia/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Radiation Chimera , Receptors, Antigen, T-Cell, alpha-beta/deficiency , Receptors, Interleukin-15/immunology , Recombinant Proteins/pharmacology , T-Lymphocyte Subsets/transplantation
18.
Sci Rep ; 11(1): 10676, 2021 05 21.
Article in English | MEDLINE | ID: mdl-34021231

ABSTRACT

The key obstacle to clinical application of human inducible regulatory T cells (iTreg) as an adoptive cell therapy in autoimmune disorders is loss of FOXP3 expression in an inflammatory milieu. Here we report human iTreg co-cultured with bone marrow-derived mesenchymal stromal cells (MSCs) during short-term ex vivo expansion enhances the stability of iTreg FOXP3 expression and suppressive function in vitro and in vivo, and further that a key mechanism of action is MSC mitochondrial (mt) transfer via tunneling nanotubules (TNT). MSC mt transfer is driven by mitochondrial metabolic function (CD39/CD73 signaling) in proliferating iTreg and promotes iTreg expression of FOXP3 stabilizing factors BACH2 and SENP3. These results elucidate cellular and molecular mechanisms underlying human MSC mt transfer to proliferating cells. MSC mt transfer stabilizes FOXP3 expression in iTregs, thereby enhancing and sustaining their suppressive function in inflammatory conditions in vitro and in vivo.


Subject(s)
Forkhead Transcription Factors/metabolism , Mesenchymal Stem Cells/metabolism , Mitochondria/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Communication , Cells, Cultured , Coculture Techniques , Cysteine Endopeptidases/metabolism , Forkhead Transcription Factors/genetics , Gene Expression , Humans , Immunomodulation , Immunophenotyping , Mice , Mitochondria/genetics , Protein Stability , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
19.
Int Immunol ; 20(9): 1201-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18632726

ABSTRACT

Enhanced basophil production is often associated with T(h)2-related conditions such as parasite infections or allergic inflammations. Our previous study demonstrated that T cell activation is necessary to promote basophil production in Nippostrongylus brasiliensis (Nb)-infected mice. Yet, mechanisms underlying how T cells aid infection-induced basophil production are not clear. In this report, we show that IL-3 produced by T cells activated by the infection enhances basophil production in Nb-infected mice. IL-3-deficient mice or Rag2-/- recipients of IL-3-deficient T cells but not of wild-type T cells failed to support basophil production following the Nb infection. Interestingly, although IL-3 was critical for preventing basophil apoptosis in vitro, IL-3 had little contribution to basophil survival and proliferation in vivo. Collectively, these results highlight a novel mechanism by which activation of adaptive immune components induces basophil production but not basophil survival via IL-3 production.


Subject(s)
Basophils/immunology , Interleukin-3/metabolism , Lymphocyte Activation/immunology , Nippostrongylus/pathogenicity , Strongylida Infections/immunology , T-Lymphocytes/immunology , Animals , Apoptosis , Basophils/physiology , Interleukin-3/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Strongylida Infections/parasitology , T-Lymphocytes/metabolism
20.
Int Immunol ; 20(2): 177-84, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18089617

ABSTRACT

The physiological functions of CD30 have not been fully elucidated. Here we show that in CD30-deficient mice (CD30(-/-)), lung inflammation is significantly diminished in the ovalbumin (OVA) model of airway hyperreactivity. In CD30(-/-) mice, the recruitment of eosinophils into the airways after OVA-aerosol challenge of OVA-primed mice was significantly diminished when compared with wild-type (w.t.) mice. IL-13 levels were also significantly reduced in CD30(-/-) mice while levels of IFN-gamma, IL-4, IL-5 and IgE in bronchoalveolar lavage fluid, lung tissue and serum were comparable to w.t. mice. Peribronchial lymph node cells from CD30(-/-) mice, re-stimulated in vitro with OVA, secreted significantly lower levels of IL-13 than those from w.t. mice, but showed normal proliferative response and other cytokine production. Exogenous IL-13 reconstituted airway recruitment of leukocytes in OVA-challenged CD3O(-/-) mice. Adoptive transfer to naive w.t. mice of in vitro OVA-re-stimulated spleen cells from CD30(-/-) mice failed to induce eosinophilic pulmonary inflammation in contrast to transfer of primed cells from w.t. mice. These results indicate that CD30 is a regulator of T(h)2 responses in the effector-memory phase and a regulator of IL-13 production in memory cells in the lung.


Subject(s)
Interleukin-13/biosynthesis , Ki-1 Antigen/metabolism , Pneumonia/immunology , Pneumonia/physiopathology , Animals , Disease Models, Animal , Eosinophils/cytology , Eosinophils/immunology , Humans , Immunologic Memory , Interleukin-13/genetics , Ki-1 Antigen/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovalbumin/administration & dosage , Ovalbumin/immunology , Th2 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL