Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 226
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 20(4): 471-481, 2019 04.
Article in English | MEDLINE | ID: mdl-30778241

ABSTRACT

Foxp3+ regulatory T cells (Treg cells) are crucial for the maintenance of immune homeostasis both in lymphoid tissues and in non-lymphoid tissues. Here we demonstrate that the ability of intestinal Treg cells to constrain microbiota-dependent interleukin (IL)-17-producing helper T cell (TH17 cell) and immunoglobulin A responses critically required expression of the transcription factor c-Maf. The terminal differentiation and function of several intestinal Treg cell populations, including RORγt+ Treg cells and follicular regulatory T cells, were c-Maf dependent. c-Maf controlled Treg cell-derived IL-10 production and prevented excessive signaling via the kinases PI(3)K (phosphatidylinositol-3-OH kinase) and Akt and the metabolic checkpoint kinase complex mTORC1 (mammalian target of rapamycin) and expression of inflammatory cytokines in intestinal Treg cells. c-Maf deficiency in Treg cells led to profound dysbiosis of the intestinal microbiota, which when transferred to germ-free mice was sufficient to induce exacerbated intestinal TH17 responses, even in a c-Maf-competent environment. Thus, c-Maf acts to preserve the identity and function of intestinal Treg cells, which is essential for the establishment of host-microbe symbiosis.


Subject(s)
Immunoglobulin A/biosynthesis , Intestines/immunology , Microbiota , Proto-Oncogene Proteins c-maf/physiology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Cells, Cultured , Colitis/immunology , Cytokines/metabolism , Dysbiosis , Gene Expression Regulation , Homeostasis , Interleukin-10/biosynthesis , Mice, Inbred C57BL , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-maf/metabolism , T-Lymphocytes, Regulatory/enzymology
2.
Cell ; 166(5): 1117-1131.e14, 2016 Aug 25.
Article in English | MEDLINE | ID: mdl-27565342

ABSTRACT

Cancer cells must evade immune responses at distant sites to establish metastases. The lung is a frequent site for metastasis. We hypothesized that lung-specific immunoregulatory mechanisms create an immunologically permissive environment for tumor colonization. We found that T-cell-intrinsic expression of the oxygen-sensing prolyl-hydroxylase (PHD) proteins is required to maintain local tolerance against innocuous antigens in the lung but powerfully licenses colonization by circulating tumor cells. PHD proteins limit pulmonary type helper (Th)-1 responses, promote CD4(+)-regulatory T (Treg) cell induction, and restrain CD8(+) T cell effector function. Tumor colonization is accompanied by PHD-protein-dependent induction of pulmonary Treg cells and suppression of IFN-γ-dependent tumor clearance. T-cell-intrinsic deletion or pharmacological inhibition of PHD proteins limits tumor colonization of the lung and improves the efficacy of adoptive cell transfer immunotherapy. Collectively, PHD proteins function in T cells to coordinate distinct immunoregulatory programs within the lung that are permissive to cancer metastasis. PAPERCLIP.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lung/immunology , Oxygen/metabolism , Prolyl Hydroxylases/metabolism , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/enzymology , Glycolysis/immunology , Interferon-gamma/immunology , Lung/pathology , Lung Neoplasms/therapy , Lymphocyte Activation , Mice , Mice, Knockout , Neoplasm Metastasis , Neuropilin-1/metabolism , Prolyl Hydroxylases/genetics , T-Lymphocytes, Regulatory/enzymology , Th1 Cells/enzymology , Th1 Cells/immunology
3.
Immunity ; 50(5): 1232-1248.e14, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31027998

ABSTRACT

Regulatory T cells (Treg cells) are important for preventing autoimmunity and maintaining tissue homeostasis, but whether Treg cells can adopt tissue- or immune-context-specific suppressive mechanisms is unclear. Here, we found that the enzyme hydroxyprostaglandin dehydrogenase (HPGD), which catabolizes prostaglandin E2 (PGE2) into the metabolite 15-keto PGE2, was highly expressed in Treg cells, particularly those in visceral adipose tissue (VAT). Nuclear receptor peroxisome proliferator-activated receptor-γ (PPARγ)-induced HPGD expression in VAT Treg cells, and consequential Treg-cell-mediated generation of 15-keto PGE2 suppressed conventional T cell activation and proliferation. Conditional deletion of Hpgd in mouse Treg cells resulted in the accumulation of functionally impaired Treg cells specifically in VAT, causing local inflammation and systemic insulin resistance. Consistent with this mechanism, humans with type 2 diabetes showed decreased HPGD expression in Treg cells. These data indicate that HPGD-mediated suppression is a tissue- and context-dependent suppressive mechanism used by Treg cells to maintain adipose tissue homeostasis.


Subject(s)
Dinoprostone/analogs & derivatives , Dinoprostone/metabolism , Hydroxyprostaglandin Dehydrogenases/metabolism , Intra-Abdominal Fat/immunology , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology , 3T3 Cells , Animals , Cell Line , Diabetes Mellitus, Type 2/metabolism , HEK293 Cells , Homeostasis/immunology , Humans , Hydroxyprostaglandin Dehydrogenases/genetics , Insulin Resistance/genetics , Intra-Abdominal Fat/cytology , Jurkat Cells , Lymphocyte Activation/immunology , Male , Mice , Mice, Knockout , STAT5 Transcription Factor/metabolism
4.
Nat Immunol ; 16(2): 188-96, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25559257

ABSTRACT

Foxp3(+) regulatory T cells (Treg cells) are required for immunological homeostasis. One notable distinction between conventional T cells (Tconv cells) and Treg cells is differences in the activity of phosphatidylinositol-3-OH kinase (PI(3)K); only Tconv cells downregulate PTEN, the main negative regulator of PI(3)K, upon activation. Here we found that control of PI(3)K in Treg cells was essential for lineage homeostasis and stability. Mice lacking Pten in Treg cells developed an autoimmune-lymphoproliferative disease characterized by excessive T helper type 1 (TH1) responses and B cell activation. Diminished control of PI(3)K activity in Treg cells led to reduced expression of the interleukin-2 (IL-2) receptor α subunit CD25, accumulation of Foxp3(+)CD25(-) cells and, ultimately, loss of expression of the transcription factor Foxp3 in these cells. Collectively, our data demonstrate that control of PI(3)K signaling by PTEN in Treg cells is critical for maintaining their homeostasis, function and stability.


Subject(s)
Homeostasis/immunology , Phosphatidylinositol 3-Kinases/metabolism , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Lineage , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Deletion , Mice , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction
5.
Nat Immunol ; 16(2): 178-87, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25559258

ABSTRACT

The interplay between effector T cells and regulatory T cells (Treg cells) is crucial for adaptive immunity, but how Treg cells control diverse effector responses is elusive. We found that the phosphatase PTEN links Treg cell stability to repression of type 1 helper T cell (TH1 cell) and follicular helper T cell (TFH cell) responses. Depletion of PTEN in Treg cells resulted in excessive TFH cell and germinal center responses and spontaneous inflammatory disease. These defects were considerably blocked by deletion of interferon-γ, indicating coordinated control of TH1 and TFH responses. Mechanistically, PTEN maintained Treg cell stability and metabolic balance between glycolysis and mitochondrial fitness. Moreover, PTEN deficiency upregulates activity of the metabolic checkpoint kinase complex mTORC2 and the serine-threonine kinase Akt, and loss of this activity restores functioning of PTEN-deficient Treg cells. Our studies establish a PTEN-mTORC2 axis that maintains Treg cell stability and coordinates Treg cell-mediated control of effector responses.


Subject(s)
PTEN Phosphohydrolase/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Animals , B-Lymphocytes/immunology , Forkhead Transcription Factors/metabolism , Humans , Lymphocyte Activation , Mice , Repressor Proteins/metabolism , Signal Transduction , Th1 Cells/enzymology
6.
Nat Immunol ; 16(3): 267-75, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25599562

ABSTRACT

The quality of the adaptive immune response depends on the differentiation of distinct CD4(+) helper T cell subsets, and the magnitude of an immune response is controlled by CD4(+)Foxp3(+) regulatory T cells (Treg cells). However, how a tissue- and cell type-specific suppressor program of Treg cells is mechanistically orchestrated has remained largely unexplored. Through the use of Treg cell-specific gene targeting, we found that the suppression of allergic immune responses in the lungs mediated by T helper type 2 (TH2) cells was dependent on the activity of the protein kinase CK2. Genetic ablation of the ß-subunit of CK2 specifically in Treg cells resulted in the proliferation of a hitherto-unexplored ILT3(+) Treg cell subpopulation that was unable to control the maturation of IRF4(+)PD-L2(+) dendritic cells required for the development of TH2 responses in vivo.


Subject(s)
Casein Kinase II/immunology , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology , Animals , CD4-Positive T-Lymphocytes/enzymology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Growth Processes/immunology , Cell Line , Dendritic Cells/enzymology , Dendritic Cells/immunology , Forkhead Transcription Factors/immunology , Humans , Hypersensitivity/blood , Hypersensitivity/immunology , Interferon Regulatory Factors/immunology , Leukocytes, Mononuclear/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Cell Surface/immunology , T-Lymphocytes, Regulatory/enzymology , Th2 Cells/enzymology
7.
Nature ; 591(7849): 306-311, 2021 03.
Article in English | MEDLINE | ID: mdl-33627871

ABSTRACT

Regulatory T cells (Treg cells) are essential for immune tolerance1, but also drive immunosuppression in the tumour microenvironment2. Therapeutic targeting of Treg cells in cancer will therefore require the identification of context-specific mechanisms that affect their function. Here we show that inhibiting lipid synthesis and metabolic signalling that are dependent on sterol-regulatory-element-binding proteins (SREBPs) in Treg cells unleashes effective antitumour immune responses without autoimmune toxicity. We find that the activity of SREBPs is upregulated in intratumoral Treg cells. Moreover, deletion of SREBP-cleavage-activating protein (SCAP)-a factor required for SREBP activity-in these cells inhibits tumour growth and boosts immunotherapy that is triggered by targeting the immune-checkpoint protein PD-1. These effects of SCAP deletion are associated with uncontrolled production of interferon-γ and impaired function of intratumoral Treg cells. Mechanistically, signalling through SCAP and SREBPs coordinates cellular programs for lipid synthesis and inhibitory receptor signalling in these cells. First, de novo fatty-acid synthesis mediated by fatty-acid synthase (FASN) contributes to functional maturation of Treg cells, and loss of FASN from Treg cells inhibits tumour growth. Second, Treg cells in tumours show enhanced expression of the PD-1 gene, through a process that depends on SREBP activity and signals via mevalonate metabolism to protein geranylgeranylation. Blocking PD-1 or SREBP signalling results in dysregulated activation of phosphatidylinositol-3-kinase in intratumoral Treg cells. Our findings show that metabolic reprogramming enforces the functional specialization of Treg cells in tumours, pointing to new ways of targeting these cells for cancer therapy.


Subject(s)
Lipid Metabolism , Neoplasms/immunology , Neoplasms/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Animals , Cholesterol/metabolism , Fatty Acid Synthases/metabolism , Fatty Acids/metabolism , Female , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/metabolism , Mevalonic Acid/metabolism , Mice , Phosphatidylinositol 3-Kinase/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Sterol Regulatory Element Binding Proteins/antagonists & inhibitors , Sterol Regulatory Element Binding Proteins/metabolism , T-Lymphocytes, Regulatory/enzymology , Up-Regulation
8.
Nature ; 565(7740): 495-499, 2019 01.
Article in English | MEDLINE | ID: mdl-30626970

ABSTRACT

Regulatory T cells (Treg cells), a distinct subset of CD4+ T cells, are necessary for the maintenance of immune self-tolerance and homeostasis1,2. Recent studies have demonstrated that Treg cells exhibit a unique metabolic profile, characterized by an increase in mitochondrial metabolism relative to other CD4+ effector subsets3,4. Furthermore, the Treg cell lineage-defining transcription factor, Foxp3, has been shown to promote respiration5,6; however, it remains unknown whether the mitochondrial respiratory chain is required for the T cell-suppression capacity, stability and survival of Treg cells. Here we report that Treg cell-specific ablation of mitochondrial respiratory chain complex III in mice results in the development of fatal inflammatory disease early in life, without affecting Treg cell number. Mice that lack mitochondrial complex III specifically in Treg cells displayed a loss of T cell-suppression capacity without altering Treg cell proliferation and survival. Treg cells deficient in complex III showed decreased expression of genes associated with Treg function, whereas Foxp3 expression remained stable. Loss of complex III in Treg cells increased DNA methylation as well as the metabolites 2-hydroxyglutarate (2-HG) and succinate that inhibit the ten-eleven translocation (TET) family of DNA demethylases7. Thus, Treg cells require mitochondrial complex III to maintain immune regulatory gene expression and suppressive function.


Subject(s)
Electron Transport Complex III/metabolism , Mitochondria/enzymology , Self Tolerance/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , DNA Demethylation , DNA Methylation , Electron Transport , Female , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Glutarates/metabolism , Inflammation/genetics , Inflammation/immunology , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Self Tolerance/genetics , Succinic Acid/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/enzymology
9.
Mol Cell ; 65(2): 296-309, 2017 Jan 19.
Article in English | MEDLINE | ID: mdl-28065600

ABSTRACT

In mammalian cells, histone deacetylase (HDAC) and Sirtuin (SIRT) are two families responsible for removing acetyl groups from acetylated proteins. Here, we describe protein deacetylation coupled with deacetylimination as a function of lysyl oxidase (LOX) family members. LOX-like 3 (Loxl3) associates with Stat3 in the nucleus to deacetylate and deacetyliminate Stat3 on multiple acetyl-lysine sites. Surprisingly, Loxl3 N-terminal scavenger receptor cysteine-rich (SRCR) repeats, rather than the C-terminal oxidase catalytic domain, represent the major deacetylase/deacetyliminase activity. Loxl3-mediated deacetylation/deacetylimination disrupts Stat3 dimerization, abolishes Stat3 transcription activity, and restricts cell proliferation. In Loxl3-/- mice, Stat3 is constitutively acetylated and naive CD4+ T cells are potentiated in Th17/Treg cell differentiation. When overexpressed, the SRCR repeats from other LOX family members can catalyze protein deacetylation/deacetylimination. Thus, our findings delineate a hitherto-unknown mechanism of protein deacetylation and deacetylimination catalyzed by lysyl oxidases.


Subject(s)
Amino Acid Oxidoreductases/metabolism , CD4-Positive T-Lymphocytes/enzymology , Colitis/enzymology , Protein Processing, Post-Translational , STAT3 Transcription Factor/metabolism , Acetylation , Amino Acid Oxidoreductases/deficiency , Amino Acid Oxidoreductases/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , Catalysis , Cell Differentiation , Cell Nucleus/enzymology , Cell Proliferation , Colitis/genetics , Colitis/immunology , Disease Models, Animal , Genotype , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Protein Domains , Protein Multimerization , RNA Interference , STAT3 Transcription Factor/genetics , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/enzymology , Th17 Cells/immunology , Transcription, Genetic , Transfection
10.
Proc Natl Acad Sci U S A ; 119(27): e2121520119, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35776543

ABSTRACT

Activated Foxp3+ regulatory T (Treg) cells differentiate into effector Treg (eTreg) cells to maintain peripheral immune homeostasis and tolerance. T cell receptor (TCR)-mediated induction and regulation of store-operated Ca2+ entry (SOCE) is essential for eTreg cell differentiation and function. However, SOCE regulation in Treg cells remains unclear. Here, we show that inositol polyphosphate multikinase (IPMK), which generates inositol tetrakisphosphate and inositol pentakisphosphate, is a pivotal regulator of Treg cell differentiation downstream of TCR signaling. IPMK is highly expressed in TCR-stimulated Treg cells and promotes a TCR-induced Treg cell program. IPMK-deficient Treg cells display aberrant T cell activation and impaired differentiation into RORγt+ Treg cells and tissue-resident Treg cells. Mechanistically, IPMK controls the generation of higher-order inositol phosphates, thereby promoting Ca2+ mobilization and Treg cell effector functions. Our findings identify IPMK as a critical regulator of TCR-mediated Ca2+ influx and highlight the importance of IPMK in Treg cell-mediated immune homeostasis.


Subject(s)
Calcium , Homeostasis , Phosphotransferases (Alcohol Group Acceptor) , Polyphosphates , T-Lymphocytes, Regulatory , Animals , Calcium/metabolism , Cell Differentiation , Homeostasis/immunology , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Polyphosphates/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology
12.
Nat Immunol ; 11(12): 1085-92, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21037577

ABSTRACT

To investigate the role of the kinase Zap70 in T cells, we generated mice expressing a Zap70 mutant whose catalytic activity can be selectively blocked by a small-molecule inhibitor. We found that conventional naive, effector and memory T cells were dependent on the kinase activity of Zap70 for their activation, which demonstrated a nonredundant role for Zap70 in signals induced by the T cell antigen receptor (TCR). In contrast, the catalytic activity of Zap70 was not required for activation of the GTPase Rap1 and inside-out signals that promote integrin adhesion. This Zap70 kinase-independent pathway was sufficient for the suppressive activity of regulatory T cells (T(reg) cells), which was unperturbed by inhibition of the catalytic activity of Zap70. Our results indicate Zap70 is a likely therapeutic target.


Subject(s)
Biocatalysis , Lymphocyte Activation/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/enzymology , ZAP-70 Protein-Tyrosine Kinase/metabolism , Animals , Cell Proliferation/drug effects , Cell Separation , Enzyme Inhibitors/pharmacology , Flow Cytometry , Immunoblotting , Immunoprecipitation , Lymphocyte Activation/drug effects , Mice , Mice, Transgenic , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/immunology
13.
J Cell Physiol ; 236(10): 7071-7087, 2021 10.
Article in English | MEDLINE | ID: mdl-33821473

ABSTRACT

Programmed death ligand 1 (PD-L1) plays a significant role in colorectal tumorigenesis through induction of regulatory T cells (Tregs) and suppression of antitumor immunity. Furthermore, microRNAs (miRNAs) as the posttranscriptional regulators of gene expression show considerable promise as a therapeutic target for colorectal cancer (CRC) treatment. Considering this, in vitro effects of miRNA-124 (miR-124-3p) on CRC cell tumorigenesis and Tregs differentiation via targeting PD-L1 were investigated in the current study. Functional analysis showed that miR-124 is significantly downregulated in CRC tissues as compared with marginal normal samples (p < .0001), and its downregulation was negatively correlated with PD-L1 expression. Moreover, a specific region in PD-L1 3'-untranslated region was predicted as the miR-124 target and validated using the luciferase assay. Further investigation showed that transfection of HT29 and SW480 cells with miR-124 mimics significantly reduced PD-L1 mRNA, protein, and cell surface expression, and inhibited Tregs in coculture models via modulating interleukin [IL]-10, IL-2, tumor necrosis factor α, transforming growth factor beta, and interferon gamma expression levels. Besides, miR-124 overexpression decreased CRC cell proliferation and arrested cell cycle at the G1 phase through downregulation of c-Myc and induced apoptosis in CRC cells via upregulation of both intrinsic and extrinsic pathways. Also, miR-124 exogenous overexpression could reduce colony and spheroid formation ability of CRC cells via downregulating CD44 mRNA expression. miR-124 also diminished MMP-9 expression and subsequently suppressed cell migration and invasion. We also illustrated that STAT3 signaling was repressed by miR-124 in CRC cells. Taken together, our findings imply that considering the involvement of miR-124 in the regulation of PD-L1 through colorectal tumorigenesis and its remarkable antitumor effects, this miRNA could be regarded as the promising target for the development of therapeutic approaches for colorectal cancer.


Subject(s)
B7-H1 Antigen/metabolism , Colorectal Neoplasms/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , MicroRNAs/metabolism , STAT3 Transcription Factor/metabolism , T-Lymphocytes, Regulatory/metabolism , Apoptosis , B7-H1 Antigen/genetics , Cell Movement , Cell Proliferation , Coculture Techniques , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Cytokines/genetics , Cytokines/metabolism , G1 Phase Cell Cycle Checkpoints , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , MicroRNAs/genetics , Neoplasm Invasiveness , STAT3 Transcription Factor/genetics , Signal Transduction , T-Lymphocytes, Regulatory/enzymology
14.
Circulation ; 142(18): 1752-1769, 2020 11 03.
Article in English | MEDLINE | ID: mdl-32900241

ABSTRACT

BACKGROUND: Hydrogen sulfide (H2S) has antihypertension and anti-inflammatory effects, and its endogenous-generation key enzyme cystathionine γ lyase (CSE) is expressed in CD4+ T cells. However, the role of CD4+ T-cell endogenous CSE/H2S in the development of hypertension is unclear. METHODS: Peripheral blood lymphocytes were isolated from hypertensive patients or spontaneously hypertensive rats, then H2S production and expression of its generation enzymes, cystathionine ß synthase and CSE, were measured to determine the major H2S generation system changes in hypertension. Mice with CSE-specific knockout in T cells (conditional knockout, by CD4cre mice hybridization) and CD4 null mice were generated for investigating the pathophysiological relevance of the CSE/H2S system. RESULTS: In lymphocytes, H2S from CSE, but not cystathionine ß synthase, responded to blood pressure changes, supported by lymphocyte CSE protein changes and a negative correlation between H2S production with systolic blood pressure and diastolic blood pressure, but positive correlation with the serum level of interleukin 10 (an anti-inflammatory cytokine). Deletion of CSE in T cells elevated BP (5-8 mm Hg) under the physiological condition and exacerbated angiotensin II-induced hypertension. In keeping with hypertension, mesenteric artery dilation impaired association with arterial inflammation, an effect attributed to reduced immunoinhibitory T regulatory cell (Treg) numbers in the blood and kidney, thus causing excess CD4+ and CD8+ T cell infiltration in perivascular adipose tissues and kidney. CSE knockout CD4+ T cell transfer into CD4 null mice, also showed the similar phenotypes' confirming the role of endogenous CSE/H2S action. Adoptive transfer of Tregs (to conditional knockout mice) reversed hypertension, vascular relaxation impairment, and immunocyte infiltration, which confirmed that conditional knockout-induced hypertension was attributable, in part, to the reduced Treg numbers. Mechanistically, endogenous CSE/H2S promoted Treg differentiation and proliferation by activating AMP-activated protein kinase. In part, it depended on activation of its upstream kinase, liver kinase B1, by sulfhydration to facilitate its substrate binding and phosphorylation. CONCLUSION: The constitutive sulfhydration of liver kinase B1 by CSE-derived H2S activates its target kinase, AMP-activated protein kinase, and promotes Treg differentiation and proliferation, which attenuates the vascular and renal immune-inflammation, thereby preventing hypertension.


Subject(s)
Cell Differentiation , Cystathionine gamma-Lyase/metabolism , Hydrogen Sulfide/metabolism , Hypertension/enzymology , Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes, Regulatory/enzymology , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases , Animals , Cystathionine gamma-Lyase/genetics , Female , Humans , Hypertension/genetics , Male , Mice , Mice, Knockout , Prospective Studies , Protein Serine-Threonine Kinases/genetics , Rats , Rats, Inbred SHR , T-Lymphocytes, Regulatory/pathology
15.
FASEB J ; 33(7): 8375-8385, 2019 07.
Article in English | MEDLINE | ID: mdl-31002527

ABSTRACT

Rheumatoid arthritis (RA) is an autoimmune disease associated with synovial hyperplasia and bone and cartilage destruction. T cells, notably T helper (Th)-1 and Th17 cells, play a critical role in the pathologic process of RA. However, it remains unclear how Th1 and Th17 cells are regulated during RA. In this study, we report that the small ubiquitin-like protein X-linked gene in the G6PD cluster at Xq28 (GdX) regulates the balance of Th17 and regulatory T (Treg) cells during collagen-induced arthritis (CIA). We discovered that the splenocytes of GdX-knockout (KO) mice were insensitive to T-cell stimulants. Correspondingly, GdX-KO mice showed alleviative Th1-mediated delayed-type hypersensitivity and were resistant to CIA compared with wild-type mice. GdX-KO mice showed fewer swollen paws, lower serum proinflammatory cytokine and anti-collagen IgG levels, and decreased synovial hyperplasia. Mechanistically, we observed that deletion of GdX decreased the transcription of proinflammatory cytokines and impaired the Th1 and Th17 differentiation but increased the Treg cell proliferation. Consistently, deletion of GdX decreased the transcription level of T-cell-specific T-box transcription factor and RAR-related orphan receptor-γ transcription factor but increased that of forkhead box P3 after being challenged with type-II collagen. These findings suggested that GdX functions as an important regulator of Th1 or Th17 and Treg cell balance during the inflammatory responses. Therefore, GdX may be a potential target for the therapy of RA.-Fu, Y., Liu, S., Wang, Y., Ren, F., Fan, X., Liang, J., Liu, C., Li, J., Ju, Y., Chang, Z. GdX/UBL4A-knockout mice resist collagen-induced arthritis by balancing the population of Th1/Th17 and regulatory T cells.


Subject(s)
Arthritis, Experimental/enzymology , T-Lymphocytes, Regulatory/enzymology , Th1 Cells/enzymology , Th17 Cells/enzymology , Ubiquitins/deficiency , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Cytokines/genetics , Cytokines/metabolism , Male , Mice , Mice, Knockout , Th1 Cells/pathology , Th17 Cells/pathology , Transcription, Genetic , Ubiquitins/metabolism
16.
Nature ; 510(7505): 407-411, 2014 06 19.
Article in English | MEDLINE | ID: mdl-24919154

ABSTRACT

Inhibitors against the p110δ isoform of phosphoinositide-3-OH kinase (PI(3)K) have shown remarkable therapeutic efficacy in some human leukaemias. As p110δ is primarily expressed in leukocytes, drugs against p110δ have not been considered for the treatment of solid tumours. Here we report that p110δ inactivation in mice protects against a broad range of cancers, including non-haematological solid tumours. We demonstrate that p110δ inactivation in regulatory T cells unleashes CD8(+) cytotoxic T cells and induces tumour regression. Thus, p110δ inhibitors can break tumour-induced immune tolerance and should be considered for wider use in oncology.


Subject(s)
Enzyme Inhibitors/pharmacology , Immune Tolerance/drug effects , Neoplasms/enzymology , Neoplasms/immunology , Phosphatidylinositol 3-Kinases/metabolism , T-Lymphocytes, Regulatory/drug effects , Animals , Antineoplastic Agents/pharmacology , Enzyme Activation/drug effects , Immune Tolerance/immunology , Mice , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology
17.
J Cell Physiol ; 234(2): 1618-1629, 2019 02.
Article in English | MEDLINE | ID: mdl-30132862

ABSTRACT

Accumulating evidence has shown that mammalian target of rapamycin (mTOR) pathway and myeloid-derived suppressor cells (MDSCs) are involved in pathogenesis of inflammatory bowel diseases (IBDs). INK128 is a novel mTOR kinase inhibitor in clinical development. However, the exact roles of MDSCs and INK128 in IBD are unclear. Here, we showed that the INK128 treatment enhanced the resistance of mice to dextran sodium sulfate (DSS)-induced colitis and inhibited the differentiation of MDSCs into macrophages. Moreover, interferon (IFN)-α level was elevated in INK128-treated colitis mice. When stimulated with IFN-α in vitro, MDSCs showed a superior immunosuppression activity. Of note, the regulatory T cells (Tregs) increased but Th1 cells decreased in INK128-treated colitis mice. These results indicate that mTOR inhibitor INK128 attenuates DSS-induced colitis via Treg expansion promoted by MDSCs. Our work provides a new evidence that INK128 is potential to be a therapeutic drug on DSS-induced colitis via regulating MDSCs as well as maintaining Treg expansion.


Subject(s)
Benzoxazoles/pharmacology , Cell Proliferation/drug effects , Colitis/prevention & control , Colon/drug effects , Lymphocyte Activation/drug effects , Myeloid-Derived Suppressor Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , T-Lymphocytes, Regulatory/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Communication/drug effects , Colitis/chemically induced , Colitis/enzymology , Colitis/immunology , Colon/enzymology , Colon/immunology , Dextran Sulfate , Disease Models, Animal , Female , Macrophages/drug effects , Macrophages/enzymology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/enzymology , Myeloid-Derived Suppressor Cells/immunology , RAW 264.7 Cells , Signal Transduction , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology , TOR Serine-Threonine Kinases/metabolism , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/metabolism
18.
Immunology ; 157(3): 210-218, 2019 07.
Article in English | MEDLINE | ID: mdl-31107985

ABSTRACT

Tumour infiltration by regulatory T (Treg) cells contributes to suppression of the anti-tumour immune response, which limits the efficacy of immune-mediated cancer therapies. The phosphoinositide 3-kinase (PI3K) pathway has key roles in mediating the function of many immune cell subsets, including Treg cells. Treg function is context-dependent and depends on input from different cell surface receptors, many of which can activate the PI3K pathway. In this review, we explore how PI3Kδ contributes to signalling through several major immune cell receptors, including the T-cell receptor and co-stimulatory receptors such as CD28 and ICOS, but is antagonized by the immune checkpoint receptors CTLA-4 and PD-1. Understanding how PI3Kδ inhibition affects Treg signalling events will help to inform how best to use PI3Kδ inhibitors in clinical cancer treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Immunotherapy , Lymphocytes, Tumor-Infiltrating/drug effects , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , T-Lymphocytes, Regulatory/drug effects , Animals , Antineoplastic Agents/adverse effects , Class Ib Phosphatidylinositol 3-Kinase/immunology , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Humans , Immunotherapy/adverse effects , Lymphocytes, Tumor-Infiltrating/enzymology , Lymphocytes, Tumor-Infiltrating/immunology , Molecular Targeted Therapy , Neoplasms/enzymology , Neoplasms/immunology , Neoplasms/pathology , Phenotype , Protein Kinase Inhibitors/adverse effects , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology , Tumor Escape , Tumor Microenvironment
19.
Ann Hematol ; 98(8): 1845-1854, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31154474

ABSTRACT

Primary immune thrombocytopenia is an autoimmune disease, characterized with decreased platelet and increased risk of bleeding. Recent studies have shown the reduction and dysfunction of regulatory T (Treg) cells in ITP patients. CD39 is highly expressed on the surface of Treg cells. It degrades ATP to AMP and CD73 dephosphorylates AMP into adenosine. Then adenosine binds with adenosine receptor and suppresses immune response by activating Treg cells and inhibiting the release of inflammatory cytokines from effector T (Teff) cells. Adenosine receptor has several subtypes and adenosine A2A receptor (A2AR) plays a crucial role especially within lymphocytes. The CD39+ Treg cells and the expression of A2AR showed abnormality in some autoimmune disease. But knowledge of CD39+ Treg cells and A2AR which are crucial in the adenosine immunosuppressive pathway is still limited in ITP. Thirty-one adult patients with newly diagnosed ITP were enrolled in this study. CD39 and A2AR expression was measured by flow cytometry and RT-PCR. The function of CD39 was reflected by the change of ATP concentration detected by CellTiter-Glo Luminescent Cell Viability Assay. CD39 expression within CD4+CD25+ Treg cells in ITP patients was decreased compared to normal controls. After high-dose dexamethasone therapy, response (R) group showed increased CD39 expression within Treg cells while non-response (NR) group did not show any difference in contrast to those before treatment. The expression of A2AR in CD4+CD25- Teff and CD4+CD25+ Treg cells was both lower in ITP patients than that of normal controls. After therapy, CD4+CD25- Teff cells had higher A2AR expression while CD4+CD25+ Treg cells did not show any difference in comparison to that before treatment. The enzymatic activity of CD39 was damaged in ITP patients and improved after high-dose dexamethasone therapy. In ITP, there was not only numerical decrease but also impaired enzymatic activity in CD39+ Treg cells. After high-dose dexamethasone treatment, these two defects could be reversed. Our results also suggested that ITP patients had reduced A2AR expression in both CD4+CD25+ Treg cells and CD4+CD25- Teff cells. CD4+CD25- Teff cells had increased A2AR expression after treatment.


Subject(s)
Apyrase/genetics , Dexamethasone/therapeutic use , Immunosuppressive Agents/therapeutic use , Purpura, Thrombocytopenic, Idiopathic/drug therapy , Receptor, Adenosine A2A/genetics , T-Lymphocytes, Regulatory/drug effects , Adenosine/immunology , Adenosine/metabolism , Adenosine Triphosphate/immunology , Adenosine Triphosphate/metabolism , Adult , Aged , Apyrase/immunology , Case-Control Studies , Female , Gene Expression , Humans , Immunophenotyping , Lymphocyte Count , Male , Middle Aged , Purpura, Thrombocytopenic, Idiopathic/enzymology , Purpura, Thrombocytopenic, Idiopathic/genetics , Purpura, Thrombocytopenic, Idiopathic/immunology , Receptor, Adenosine A2A/immunology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/enzymology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology
20.
Immunity ; 32(5): 670-80, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20493730

ABSTRACT

T cell activation is tightly regulated to avoid autoimmunity. Gene related to anergy in lymphocytes (GRAIL, encoded by Rnf128) is an E3 ubiquitin ligase associated with T cell tolerance. Here, we generated and analyzed GRAIL-deficient mice and found they were resistant to immune tolerance induction and exhibited greater susceptibility to autoimmune diseases than wild-type mice. GRAIL-deficient naive T cells, after activation, exhibited increased proliferation and cytokine expression than controls and did not depend on costimulation for effector generation. Moreover, GRAIL-deficient regulatory T (Treg) cells displayed reduced suppressive function, associated with increased Th17 cell-related gene expression. GRAIL-deficient naive and Treg cells were less efficient in downregulating T cell receptor (TCR)-CD3 expression after activation and exhibited increased NFATc1 transcription factor expression; GRAIL expression promoted CD3 ubiquitinylation. Our results indicate that GRAIL, by mediating TCR-CD3 degradation, regulates naive T cell tolerance induction and Treg cell function.


Subject(s)
CD3 Complex/immunology , Immune Tolerance/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/immunology , Ubiquitin-Protein Ligases/immunology , Animals , Gene Expression Regulation , Immunoblotting , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , T-Lymphocytes, Regulatory/enzymology , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL