Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
1.
Nature ; 617(7960): 377-385, 2023 05.
Article in English | MEDLINE | ID: mdl-37138075

ABSTRACT

The gut microbiota is a crucial regulator of anti-tumour immunity during immune checkpoint inhibitor therapy. Several bacteria that promote an anti-tumour response to immune checkpoint inhibitors have been identified in mice1-6. Moreover, transplantation of faecal specimens from responders can improve the efficacy of anti-PD-1 therapy in patients with melanoma7,8. However, the increased efficacy from faecal transplants is variable and how gut bacteria promote anti-tumour immunity remains unclear. Here we show that the gut microbiome downregulates PD-L2 expression and its binding partner repulsive guidance molecule b (RGMb) to promote anti-tumour immunity and identify bacterial species that mediate this effect. PD-L1 and PD-L2 share PD-1 as a binding partner, but PD-L2 can also bind RGMb. We demonstrate that blockade of PD-L2-RGMb interactions can overcome microbiome-dependent resistance to PD-1 pathway inhibitors. Antibody-mediated blockade of the PD-L2-RGMb pathway or conditional deletion of RGMb in T cells combined with an anti-PD-1 or anti-PD-L1 antibody promotes anti-tumour responses in multiple mouse tumour models that do not respond to anti-PD-1 or anti-PD-L1 alone (germ-free mice, antibiotic-treated mice and even mice colonized with stool samples from a patient who did not respond to treatment). These studies identify downregulation of the PD-L2-RGMb pathway as a specific mechanism by which the gut microbiota can promote responses to PD-1 checkpoint blockade. The results also define a potentially effective immunological strategy for treating patients who do not respond to PD-1 cancer immunotherapy.


Subject(s)
Drug Resistance, Neoplasm , Immunotherapy , Melanoma , Microbiota , Animals , Humans , Mice , Cell Adhesion Molecules, Neuronal , Disease Models, Animal , Down-Regulation , Drug Resistance, Neoplasm/drug effects , Fecal Microbiota Transplantation , Germ-Free Life , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Melanoma/immunology , Melanoma/microbiology , Melanoma/therapy , Protein Binding/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
2.
Nat Immunol ; 15(6): 562-70, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24777531

ABSTRACT

Deubiquitinases (DUBs) are a new class of drug targets, although the physiological function of only few DUBs has been characterized. Here we identified the DUB USP15 as a crucial negative regulator of T cell activation. USP15 stabilized the E3 ubiquitin ligase MDM2, which in turn negatively regulated T cell activation by targeting the degradation of the transcription factor NFATc2. USP15 deficiency promoted T cell activation in vitro and enhanced T cell responses to bacterial infection and tumor challenge in vivo. USP15 also stabilized MDM2 in cancer cells and regulated p53 function and cancer-cell survival. Our results suggest that inhibition of USP15 may both induce tumor cell apoptosis and boost antitumor T cell responses.


Subject(s)
NFATC Transcription Factors/metabolism , Proto-Oncogene Proteins c-mdm2/immunology , Th1 Cells/immunology , Ubiquitin-Specific Proteases/immunology , Adoptive Transfer , Animals , Apoptosis/immunology , Cell Differentiation/immunology , Cell Line, Tumor , Cell Survival , HCT116 Cells , Humans , Leupeptins/pharmacology , Listeria monocytogenes/immunology , Listeriosis/immunology , Lymphocyte Activation/immunology , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-mdm2/genetics , Tumor Escape , Tumor Suppressor Protein p53/immunology , Ubiquitin-Specific Proteases/genetics , Ubiquitination/genetics , Ubiquitination/immunology
3.
Nat Immunol ; 14(7): 732-40, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23727894

ABSTRACT

Transcription factors of the STAT family are critical in the cytokine-mediated functional differentiation of CD4(+) helper T cells. Signaling inhibitors of the SOCS family negatively regulate the activation of STAT proteins; however, their roles in the differentiation and function of helper T cells are not well understood. Here we found that the SOCS protein CIS, which was substantially induced by interleukin 4 (IL-4), negatively regulated the activation of STAT3, STAT5 and STAT6 in T cells. CIS-deficient mice spontaneously developed airway inflammation, and CIS deficiency in T cells led to greater susceptibility to experimental allergic asthma. CIS-deficient T cells showed enhanced differentiation into the TH2 and TH9 subsets of helper T cells. STAT5 and STAT6 regulated IL-9 expression by directly binding to the Il9 promoter. Our data thus demonstrate a critical role for CIS in controlling the proallergic generation of helper T cells.


Subject(s)
Asthma/immunology , Inflammation/immunology , STAT Transcription Factors/immunology , Suppressor of Cytokine Signaling Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation/immunology , Histocytochemistry , Immunoblotting , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , RNA/chemistry , RNA/genetics , Real-Time Polymerase Chain Reaction , Signal Transduction , Suppressor of Cytokine Signaling Proteins/genetics
4.
J Immunol ; 209(7): 1286-1299, 2022 10 01.
Article in English | MEDLINE | ID: mdl-36038291

ABSTRACT

Type I conventional dendritic cells (cDC1s) are an essential Ag-presenting population required for generating adaptive immunity against intracellular pathogens and tumors. While the transcriptional control of cDC1 development is well understood, the mechanisms by which extracellular stimuli regulate cDC1 function remain unclear. We previously demonstrated that the cytokine-responsive transcriptional regulator STAT3 inhibits polyinosinic:polycytidylic acid [poly(I:C)]-induced cDC1 maturation and cDC1-mediated antitumor immunity in murine breast cancer, indicating an intrinsic, suppressive role for STAT3 in cDC1s. To probe transcriptional mechanisms regulating cDC1 function, we generated novel RNA sequencing datasets representing poly(I:C)-, IL-10-, and STAT3-mediated gene expression responses in murine cDC1s. Bioinformatics analyses indicated that poly(I:C) stimulates multiple inflammatory pathways independent of STAT3, while IL-10-activated STAT3 uniquely inhibits the poly(I:C)-induced type I IFN (IFN-I) transcriptional response. We validated this mechanism using purified cDC1s deficient for STAT3 or IFN signaling. Our data reveal IL-10-activated STAT3 suppresses production of IFN-ß and IFN-γ, accrual of tyrosine phosphorylated STAT1, and IFN-stimulated gene expression in cDC1s after poly(I:C) exposure. Moreover, we found that maturation of cDC1s in response to poly(I:C) is dependent on the IFN-I receptor, but not the type II IFN receptor, or IFN-λ. Taken together, we elucidate an essential role for STAT3 in restraining autocrine IFN-I signaling in cDC1s elicited by poly(I:C) stimulation, and we provide novel RNA sequencing datasets that will aid in further delineating inflammatory and anti-inflammatory mechanisms in cDC1s.


Subject(s)
Interleukin-10 , STAT3 Transcription Factor , Animals , Cytokines/metabolism , Dendritic Cells , Interleukin-10/metabolism , Mice , Poly I-C/pharmacology , STAT3 Transcription Factor/metabolism , Tyrosine/metabolism
6.
Nat Immunol ; 12(12): 1221-9, 2011 Nov 06.
Article in English | MEDLINE | ID: mdl-22057289

ABSTRACT

During infection, naive CD8(+) T cells differentiate into effector cells, which are armed to eliminate pathogens, and memory cells, which are poised to protect against reinfection. The transcriptional program that regulates terminal differentiation into short-lived effector-memory versus long-lived memory cells is not clearly defined. Through the use of mice expressing reporters for the DNA-binding inhibitors Id2 and Id3, we identified Id3(hi) precursors of long-lived memory cells before the peak of T cell population expansion or upregulation of cell-surface receptors that indicate memory potential. Deficiency in Id2 or Id3 resulted in loss of distinct CD8(+) effector and memory populations, which demonstrated unique roles for these inhibitors of E-protein transcription factors. Furthermore, cytokines altered the expression of Id2 and Id3 differently, which provides insight into how external cues influence gene expression.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Gene Expression Regulation , Immunologic Memory/immunology , Inhibitor of Differentiation Protein 2/metabolism , Inhibitor of Differentiation Proteins/metabolism , T-Lymphocyte Subsets/immunology , Transcription, Genetic , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Cytokines/pharmacology , Gene Expression Regulation/drug effects , Infections/genetics , Infections/immunology , Infections/microbiology , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Proteins/genetics , Lectins, C-Type , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Receptors, Immunologic/metabolism , T-Lymphocyte Subsets/cytology , Transcription, Genetic/drug effects
7.
Immunity ; 40(3): 342-54, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24656046

ABSTRACT

Production of type I interferons (IFN-I) is a crucial innate immune mechanism against viral infections. IFN-I induction is subject to negative regulation by both viral and cellular factors, but the underlying mechanism remains unclear. We report that the noncanonical NF-κB pathway was stimulated along with innate immune cell differentiation and viral infections and had a vital role in negatively regulating IFN-I induction. Genetic deficiencies in major components of the noncanonical NF-κB pathway caused IFN-I hyperinduction and rendered cells and mice substantially more resistant to viral infection. Noncanonical NF-κB suppressed signal-induced histone modifications at the Ifnb promoter, an action that involved attenuated recruitment of the transcription factor RelA and a histone demethylase, JMJD2A. These findings reveal an unexpected function of the noncanonical NF-κB pathway and highlight an important mechanism regulating antiviral innate immunity.


Subject(s)
Immunity, Innate , Interferon Type I/biosynthesis , NF-kappa B/metabolism , Virus Diseases/immunology , Virus Diseases/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Enzyme Activation , Female , Gene Expression Regulation/drug effects , Hematopoietic Cell Growth Factors/pharmacology , Histone Demethylases/metabolism , Histones/metabolism , Immunity, Innate/drug effects , Interferon-beta/genetics , Interferon-beta/metabolism , Ligands , Mice , Promoter Regions, Genetic , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Toll-Like Receptors/metabolism , Transcription Factor RelA/metabolism , Virus Diseases/genetics , NF-kappaB-Inducing Kinase
8.
Proc Natl Acad Sci U S A ; 115(10): E2311-E2319, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29463696

ABSTRACT

Blood cell formation must be appropriately maintained throughout life to provide robust immune function, hemostasis, and oxygen delivery to tissues, and to prevent disorders that result from over- or underproduction of critical lineages. Persistent inflammation deregulates hematopoiesis by damaging hematopoietic stem and progenitor cells (HSPCs), leading to elevated myeloid cell output and eventual bone marrow failure. Nonetheless, antiinflammatory mechanisms that protect the hematopoietic system are understudied. The transcriptional regulator STAT3 has myriad roles in HSPC-derived populations and nonhematopoietic tissues, including a potent antiinflammatory function in differentiated myeloid cells. STAT3 antiinflammatory activity is facilitated by STAT3-mediated transcriptional repression of Ube2n, which encodes the E2 ubiquitin-conjugating enzyme Ubc13 involved in proinflammatory signaling. Here we demonstrate a crucial role for STAT3 antiinflammatory activity in preservation of HSPCs and lineage-balanced hematopoiesis. Conditional Stat3 removal from the hematopoietic system led to depletion of the bone marrow lineage- Sca-1+ c-Kit+ CD150+ CD48- HSPC subset (LSK CD150+ CD48- cells), myeloid-skewed hematopoiesis, and accrual of DNA damage in HSPCs. These responses were accompanied by intrinsic transcriptional alterations in HSPCs, including deregulation of inflammatory, survival and developmental pathways. Concomitant Ube2n/Ubc13 deletion from Stat3-deficient hematopoietic cells enabled lineage-balanced hematopoiesis, mitigated depletion of bone marrow LSK CD150+ CD48- cells, alleviated HSPC DNA damage, and corrected a majority of aberrant transcriptional responses. These results indicate an intrinsic protective role for STAT3 in the hematopoietic system, and suggest that this is mediated by STAT3-dependent restraint of excessive proinflammatory signaling via Ubc13 modulation.


Subject(s)
Blood Cells/immunology , Hematopoiesis , STAT3 Transcription Factor/immunology , Animals , Blood Cells/cytology , Cell Lineage , Female , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Male , Mice , Mice, Inbred C57BL , Myeloid Cells/cytology , Myeloid Cells/immunology , STAT3 Transcription Factor/genetics , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/immunology
9.
Carcinogenesis ; 41(11): 1529-1542, 2020 11 13.
Article in English | MEDLINE | ID: mdl-32603404

ABSTRACT

K-ras mutant lung adenocarcinoma (LUAD) is the most common type of lung cancer, displays abysmal prognosis and is tightly linked to tumor-promoting inflammation, which is increasingly recognized as a target for therapeutic intervention. We have recently shown a gender-specific role for epithelial Stat3 signaling in the pathogenesis of K-ras mutant LUAD. The absence of epithelial Stat3 in male K-ras mutant mice (LR/Stat3Δ/Δ mice) promoted tumorigenesis and induced a nuclear factor-kappaB (NF-κB)-driven pro-tumor immune response while reducing tumorigenesis and enhancing anti-tumor immunity in female counterparts. In the present study, we manipulated estrogen and NF-κB signaling to study the mechanisms underlying this intriguing gender-disparity. In LR/Stat3Δ/Δ females, estrogen deprivation by bilateral oophorectomy resulted in higher tumor burden, an induction of NF-κB-driven immunosuppressive response, and reduced anti-tumor cytotoxicity, whereas estrogen replacement reversed these changes. On the other hand, exogenous estrogen in males successfully inhibited tumorigenesis, attenuated NF-κB-driven immunosuppression and boosted anti-tumor immunity. Mechanistically, genetic targeting of epithelial NF-κB activity resulted in reduced tumorigenesis and enhanced the anti-tumor immune response in LR/Stat3Δ/Δ males, but not females. Our data suggest that estrogen exerts a context-specific anti-tumor effect through inhibiting NF-κB-driven tumor-promoting inflammation and provide insights into developing novel personalized therapeutic strategies for K-ras mutant LUAD.


Subject(s)
Adenocarcinoma of Lung/immunology , Cell Transformation, Neoplastic/immunology , Estrogens/metabolism , Immunomodulation , Lung Neoplasms/immunology , NF-kappa B/metabolism , STAT3 Transcription Factor/metabolism , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Animals , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Female , Humans , Immunity/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Mutation , NF-kappa B/genetics , Proto-Oncogene Proteins p21(ras)/genetics , STAT3 Transcription Factor/genetics , Tumor Cells, Cultured
10.
Gastroenterology ; 157(1): 163-178, 2019 07.
Article in English | MEDLINE | ID: mdl-30885780

ABSTRACT

BACKGROUND & AIMS: The peroxisome proliferator-activated receptor delta (PPARD) regulates cell metabolism, proliferation, and inflammation and has been associated with gastric and other cancers. Villin-positive epithelial cells are a small population of quiescent gastric progenitor cells. We expressed PPARD from a villin promoter to investigate the role of these cells and PPARD in development of gastric cancer. METHODS: We analyzed gastric tissues from mice that express the Ppard (PPARD1 and PPARD2 mice) from a villin promoter, and mice that did not carry this transgene (controls), by histology and immunohistochemistry. We performed cell lineage-tracing experiments and analyzed the microbiomes, chemokine and cytokine production, and immune cells and transcriptomes of stomachs of these mice. We also performed immunohistochemical analysis of PPARD levels in 2 sets of human gastric tissue microarrays. RESULTS: Thirty-eight percent of PPARD mice developed spontaneous, invasive gastric adenocarcinomas, with severe chronic inflammation. Levels of PPARD were increased in human gastric cancer tissues, compared with nontumor tissues, and associated with gastric cancer stage and grade. We found an inverse correlation between level of PPARD in tumor tissue and patient survival time. Gastric microbiomes from PPARD and control mice did not differ significantly. Lineage-tracing experiments identified villin-expressing gastric progenitor cells (VGPCs) as the origin of gastric tumors in PPARD mice. In these mice, PPARD up-regulated CCL20 and CXCL1, which increased infiltration of the gastric mucosa by immune cells. Immune cell production of inflammatory cytokines promoted chronic gastric inflammation and expansion and transformation of VGPCs, leading to tumorigenesis. We identified a positive-feedback loop between PPARD and interferon gamma signaling that sustained gastric inflammation to induce VGPC transformation and gastric carcinogenesis. CONCLUSIONS: We found PPARD overexpression in VPGCs to result in inflammation, dysplasia, and tumor formation. PPARD and VGPCs might be therapeutic targets for stomach cancer.


Subject(s)
Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics , Cytokines/immunology , Gastric Mucosa/metabolism , Interferon-gamma/immunology , Receptors, Cytoplasmic and Nuclear/genetics , Stem Cells/metabolism , Stomach/immunology , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Animals , Carcinogenesis/immunology , Cell Lineage , Cell Transformation, Neoplastic/immunology , Chemokine CCL20/metabolism , Chemokine CXCL1/metabolism , Chemokines , Feedback, Physiological , Gene Expression Profiling , Inflammation , Mice , Microbiota/immunology , Microfilament Proteins/genetics , Stem Cells/immunology , Stomach/microbiology , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology
11.
Immunity ; 30(4): 576-87, 2009 Apr 17.
Article in English | MEDLINE | ID: mdl-19362022

ABSTRACT

T helper (Th) 17 cells have been recently discovered in both mouse and human. Here we show that interleukin-1 (IL-1) signaling on T cells is critically required for the early programming of Th17 cell lineage and Th17 cell-mediated autoimmunity. IL-1 receptor1 expression in T cells, which was induced by IL-6, was necessary for the induction of experimental autoimmune encephalomyelitis and for early Th17 cell differentiation in vivo. Moreover, IL-1 signaling in T cells was required in dendritic cell-mediated Th17 cell differentiation from naive or regulatory precursors and IL-1 synergized with IL-6 and IL-23 to regulate Th17 cell differentiation and maintain cytokine expression in effector Th17 cells. Importantly, IL-1 regulated the expression of the transcription factors IRF4 and RORgammat during Th17 cell differentiation; overexpression of these two factors resulted in IL-1-independent Th17 cell polarization. Our data thus indicate a critical role of IL-1 in Th17 cell differentiation and this pathway may serve as a unique target for Th17 cell-mediated immunopathology.


Subject(s)
Cell Differentiation , Gene Expression Regulation , Interleukin-1/metabolism , Signal Transduction , T-Lymphocyte Subsets/cytology , T-Lymphocytes, Helper-Inducer/cytology , Animals , Cell Lineage , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , RNA, Messenger/biosynthesis , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , Up-Regulation
12.
Immunol Rev ; 261(1): 84-101, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25123278

ABSTRACT

The term innate immunity typically refers to a quick but non-specific host defense response against invading pathogens. The innate immune system comprises particular immune cell populations, epithelial barriers, and numerous secretory mediators including cytokines, chemokines, and defense peptides. Innate immune cells are also now recognized to play important contributing roles in cancer and pathological inflammatory conditions. Innate immunity relies on rapid signal transduction elicited upon pathogen recognition via pattern recognition receptors (PRRs) and cell:cell communication conducted by soluble mediators, including cytokines. A majority of cytokines involved in innate immune signaling use a molecular cascade encompassing receptor-associated Jak protein tyrosine kinases and STAT (signal transducer and activator of transcription) transcriptional regulators. Here, we focus on roles for STAT proteins in three major innate immune subsets: neutrophils, macrophages, and dendritic cells (DCs). While knowledge in this area is only now emerging, understanding the molecular regulation of these cell types is necessary for developing new approaches to treat human disorders such as inflammatory conditions, autoimmunity, and cancer.


Subject(s)
Dendritic Cells/immunology , Immune System Diseases/immunology , Macrophages/immunology , Neoplasms/immunology , Neutrophils/immunology , STAT Transcription Factors/metabolism , Animals , Cell Communication/immunology , Cytokines/metabolism , Humans , Immunity, Innate , Janus Kinases/metabolism , Receptors, Pattern Recognition/metabolism , STAT Transcription Factors/genetics , Signal Transduction , Transcriptional Activation
13.
Immunity ; 28(4): 509-20, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18342552

ABSTRACT

The development of distinct dendritic cell (DC) subsets is regulated by cytokines. The ligand for the FMS-like tyrosine kinase 3 receptor (Flt3L) is necessary for plasmacytoid DC (pDC) and conventional DC (cDC) maturation. The cytokine GM-CSF inhibits Flt3L-driven pDC production while promoting cDC growth. We show that GM-CSF selectively utilized its signal transducer STAT5 to block Flt3L-dependent pDC development from the lineage-negative, Flt3+ (lin- Flt3+) bone-marrow subset. The signaling molecule STAT3, by contrast, was necessary for expansion of DC progenitors but not pDC maturation. In vivo, STAT5 suppressed pDC formation during repopulation of the DC compartment after bone-marrow ablation. GM-CSF-dependent STAT5 signaling rapidly extinguished pDC-related gene expression in lin- Flt3+ progenitors. Inspection of the Irf8 promoter revealed that STAT5 was recruited during GM-CSF-mediated suppression, indicating that STAT5 directly inhibited transcription of this critical pDC gene. Our results therefore show that GM-CSF controls the production of pDCs by employing STAT5 to suppress IRF8 and the pDC transcriptional network in lin- Flt3+ progenitors.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Growth Inhibitors/physiology , Interferon Regulatory Factors/antagonists & inhibitors , STAT5 Transcription Factor/physiology , Signal Transduction/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Lineage/immunology , Cells, Cultured , Dendritic Cells/cytology , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Interferon Regulatory Factors/biosynthesis , Interferon Regulatory Factors/physiology , Mice , Mice, Knockout , Multipotent Stem Cells/cytology , Multipotent Stem Cells/immunology , Multipotent Stem Cells/metabolism , STAT5 Transcription Factor/deficiency , STAT5 Transcription Factor/genetics , Signal Transduction/genetics , fms-Like Tyrosine Kinase 3/biosynthesis
14.
Immunity ; 29(1): 138-49, 2008 Jul 18.
Article in English | MEDLINE | ID: mdl-18599325

ABSTRACT

After activation, CD4(+) helper T (Th) cells differentiate into distinct effector subsets. Although chemokine (C-X-C motif) receptor 5-expressing T follicular helper (Tfh) cells are important in humoral immunity, their developmental regulation is unclear. Here we show that Tfh cells had a distinct gene expression profile and developed in vivo independently of the Th1 or Th2 cell lineages. Tfh cell generation was regulated by ICOS ligand (ICOSL) expressed on B cells and was dependent on interleukin-21 (IL-21), IL-6, and signal transducer and activator of transcription 3 (STAT3). However, unlike Th17 cells, differentiation of Tfh cells did not require transforming growth factor beta (TGF-beta) or Th17-specific orphan nuclear receptors RORalpha and RORgamma in vivo. Finally, naive T cells activated in vitro in the presence of IL-21 but not TGF-beta signaling preferentially acquired Tfh gene expression and promoted germinal-center reactions in vivo. This study thus demonstrates that Tfh is a distinct Th cell lineage.


Subject(s)
Cell Differentiation/immunology , Cell Lineage/immunology , Germinal Center/cytology , Interleukins/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocytes, Helper-Inducer/cytology , Adoptive Transfer , Animals , B-Lymphocytes/immunology , Flow Cytometry , Gene Expression , Gene Expression Profiling , Germinal Center/immunology , Immunohistochemistry , Inducible T-Cell Co-Stimulator Ligand , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Interleukins/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Mutant Strains , Oligonucleotide Array Sequence Analysis , Proteins/immunology , Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism
15.
Immunity ; 29(1): 44-56, 2008 Jul 18.
Article in English | MEDLINE | ID: mdl-18585065

ABSTRACT

Regulatory T (Treg) and T helper 17 (Th17) cells were recently proposed to be reciprocally regulated during differentiation. To understand the underlying mechanisms, we utilized a Th17 reporter mouse with a red fluorescent protein (RFP) sequence inserted into the interleukin-17F (IL-17F) gene. Using IL-17F-RFP together with a Foxp3 reporter, we found that the development of Th17 and Foxp3(+) Treg cells was associated in immune responses. Although TGF-beta receptor I signaling was required for both Foxp3 and IL-17 induction, SMAD4 was only involved in Foxp3 upregulation. Foxp3 inhibited Th17 differentiation by antagonizing the function of the transcription factors RORgammat and ROR*. In contrast, IL-6 overcame this suppressive effect of Foxp3 and, together with IL-1, induced genetic reprogramming in Foxp3(+) Treg cells. STAT3 regulated Foxp3 downregulation, whereas STAT3, RORgamma, and ROR* were required for IL-17 expression in Treg cells. Our data demonstrate molecular antagonism and plasticity of Treg and Th17 cell programs.


Subject(s)
Cell Differentiation/immunology , Inflammation/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Regulatory/cytology , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Flow Cytometry , Immunoprecipitation , Interleukin-17/immunology , Interleukin-17/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Polymerase Chain Reaction , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Transduction, Genetic , Transforming Growth Factor beta1/immunology , Transforming Growth Factor beta1/metabolism
16.
Blood ; 124(3): 403-11, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-24891320

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) is considered a negative regulator of inflammation, as inhibition of STAT3 signaling enhances antitumor immunity. However, STAT3 activation is a key oncogenic pathway in natural killer (NK)-lineage large granular lymphomas, and we recently reported enhanced proliferation and function of human NK cells activated with IL-21, which signals primarily through STAT3. These IL-21-expanded NK cells also have increased NKG2D expression, which led us to focus our investigation on whether STAT3 regulates NKG2D. In this study, we show that modulation of STAT3 phosphorylation with cytokines and small-molecule inhibitors correlates with NKG2D expression on human NK cells, leading to altered NK-cell degranulation. Moreover, NKG2D expression on murine NK cells having conditional STAT3 ablation is lower than on NK cells from wild-type mice, and human NK cells carrying dominant-negative STAT3 mutations have decreased baseline NKG2D expression and blunted responses to IL-10 and IL-21. Lastly, we show binding of STAT3 to a predicted STAT3 binding site upstream of the NKG2D gene, which is enhanced by IL-10 and IL-21 and decreased by STAT3 inhibition. Taken together, these data show that NKG2D expression in NK cells is regulated at the transcriptional level by STAT3, resulting in a functional NK cell defect in patients with STAT3 mutations.


Subject(s)
Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , NK Cell Lectin-Like Receptor Subfamily K/genetics , STAT3 Transcription Factor/metabolism , Animals , Base Sequence , Binding Sites/genetics , DNA/genetics , DNA/metabolism , Humans , Interleukin-10/metabolism , Interleukin-15/metabolism , Interleukins/metabolism , Job Syndrome/genetics , Job Syndrome/immunology , Job Syndrome/metabolism , Mice , Mice, Knockout , Molecular Sequence Data , Mutation , Phosphorylation , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/deficiency , STAT3 Transcription Factor/genetics , Signal Transduction , Transcription, Genetic , Tyrosine/metabolism
17.
FASEB J ; 29(6): 2359-70, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25713055

ABSTRACT

The IL-6/signal transducer and activator of transcription 3 (STAT3) pathway is a critical signaling pathway for colitis-associated colorectal cancer (CAC). Peroxisome proliferator-activated receptor (PPAR)-δ, a lipid nuclear receptor, up-regulates IL-6. 15-Lipoxygenase-1 (15-LOX-1), which is crucial to production of lipid signaling mediators to terminate inflammation, down-regulates PPAR-δ. 15-LOX-1 effects on IL-6/STAT3 signaling and CAC tumorigenesis have not been determined. We report that intestinally targeted transgenic 15-LOX-1 expression in mice inhibited azoxymethane- and dextran sodium sulfate-induced CAC, IL-6 expression, STAT3 phosphorylation, and IL-6/STAT3 downstream target (Notch3 and MUC1) expression. 15-LOX-1 down-regulation was associated with IL-6 up-regulation in human colon cancer mucosa. Reexpression of 15-LOX-1 in human colon cancer cells suppressed IL-6 mRNA expression, STAT3 phosphorylation, IL-6 promoter activity, and PPAR-δ mRNA and protein expression. PPAR-δ overexpression in colonic epithelial cells promoted CAC tumorigenesis in mice and increased IL-6 expression and STAT3 phosphorylation, whereas concomitant 15-LOX-1 expression in colonic epithelial cells (15-LOX-1-PPAR-δ-Gut mice) suppressed these effects: the number of tumors per mouse (mean ± sem) was 4.22 ± 0.68 in wild-type littermates, 6.67 ± 0.83 in PPAR-δ-Gut mice (P = 0.026), and 2.25 ± 0.25 in 15-LOX-1-PPAR-δ-Gut mice (P = 0.0006). Identification of 15-LOX-1 suppression of PPAR-δ to inhibit IL-6/STAT3 signaling-driven CAC tumorigenesis provides mechanistic insights that can be used to molecularly target CAC.


Subject(s)
Arachidonate 15-Lipoxygenase/metabolism , Colitis/metabolism , Colonic Neoplasms/metabolism , Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism , Animals , Arachidonate 15-Lipoxygenase/genetics , Azoxymethane , Blotting, Western , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation/genetics , Colitis/chemically induced , Colitis/genetics , Colonic Neoplasms/chemically induced , Colonic Neoplasms/genetics , Dextran Sulfate , Gene Expression , HCT116 Cells , Humans , Immunohistochemistry , Interleukin-6/genetics , Mice, Transgenic , PPAR delta/genetics , PPAR delta/metabolism , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics
18.
Eur J Haematol ; 97(3): 261-70, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26660446

ABSTRACT

Heterozygous mutations in the transcriptional regulator GATA-2 associate with multilineage immunodeficiency, myelodysplastic syndrome (MDS), and acute myeloid leukemia (AML). The majority of these mutations localize in the zinc finger (ZnF) domains, which mediate GATA-2 DNA binding. Deregulated hematopoiesis with GATA-2 mutation frequently develops in adulthood, yet GATA-2 function in the bone marrow remains unresolved. To investigate this, we conditionally deleted the GATA-2 C-terminal ZnF (C-ZnF) coding sequences in adult mice. Upon Gata2 C-ZnF deletion, we observed rapid peripheral cytopenia, bone marrow failure, and decreased c-Kit expression on hematopoietic progenitors. Transplant studies indicated GATA-2 has a cell-autonomous role in bone marrow hematopoiesis. Moreover, myeloid lineage populations were particularly sensitive to Gata2 hemizygosity, while molecular assays indicated GATA-2 regulates c-Kit expression in multilineage progenitor cells. Enforced c-Kit expression in Gata2 C-ZnF-deficient hematopoietic progenitors enhanced myeloid colony activity, suggesting GATA-2 sustains myelopoiesis via a cell intrinsic role involving maintenance of c-Kit expression. Our results provide insight into mechanisms regulating hematopoiesis in bone marrow and may contribute to a better understanding of immunodeficiency and bone marrow failure associated with GATA-2 mutation.


Subject(s)
Anemia, Aplastic/genetics , Bone Marrow Diseases/genetics , Bone Marrow/pathology , GATA2 Transcription Factor/genetics , Hemoglobinuria, Paroxysmal/genetics , Protein Interaction Domains and Motifs/genetics , Proto-Oncogene Proteins c-kit/deficiency , Sequence Deletion , Zinc Fingers/genetics , Anemia, Aplastic/diagnosis , Anemia, Aplastic/metabolism , Anemia, Aplastic/mortality , Animals , Biomarkers , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Marrow Diseases/diagnosis , Bone Marrow Diseases/metabolism , Bone Marrow Diseases/mortality , Bone Marrow Failure Disorders , Bone and Bones/pathology , Chromatin Immunoprecipitation , Decalcification, Pathologic/genetics , Disease Models, Animal , GATA2 Transcription Factor/chemistry , GATA2 Transcription Factor/metabolism , Gene Expression , Gene Expression Regulation , Gene Frequency , Genes, Reporter , Genotype , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Hemoglobinuria, Paroxysmal/diagnosis , Hemoglobinuria, Paroxysmal/metabolism , Hemoglobinuria, Paroxysmal/mortality , High-Throughput Nucleotide Sequencing , Immunophenotyping , Mice , Mice, Knockout , Prognosis , Side-Population Cells
19.
Oncologist ; 19(5): 536-44, 2014 May.
Article in English | MEDLINE | ID: mdl-24705981

ABSTRACT

The Janus kinase (JAK) and signal transducer and activator of transcription (STAT) pathway is an active mediator of cytokine signaling in the pathogenesis of solid and hematologic malignancies. The seven-member STAT family is composed of latent cytoplasmic transcription factors that are activated by phosphorylation intertwined in a network with activation that ultimately leads to cell proliferation. An activated kinase enzyme phosphorylates one STAT factor or more, which shuttle to the nucleus to regulate gene expression, promoting cell survival. Somatic STAT3 mutations have been recently reported in large granular lymphocytic leukemia, aplastic anemia, and myelodysplastic syndrome. Furthermore, the relationship between BCL6 and STAT3 in diffuse large B-cell lymphomas, particularly on the activated B-cell subtype, needs to be further explored. The search for therapeutic STAT3 inhibitors that abrogate the JAK/STAT pathway is currently under way. Targeting the STAT pathway, which seems to be critical in tumorigenesis, is promising for multiple malignancies including lymphoma and leukemia. In this paper, we review mechanisms of action, failures, and successes of STAT3 inhibitors.


Subject(s)
Leukemia/drug therapy , Lymphoma/drug therapy , STAT3 Transcription Factor/antagonists & inhibitors , Cell Proliferation , Cell Survival/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Interleukin-6/antagonists & inhibitors , Interleukin-6/metabolism , Janus Kinases/antagonists & inhibitors , Phosphorylation , Proto-Oncogene Proteins c-bcl-6 , STAT3 Transcription Factor/genetics
20.
Blood ; 120(22): 4363-73, 2012 Nov 22.
Article in English | MEDLINE | ID: mdl-23033267

ABSTRACT

Cytokines and transcription factors play key roles in dendritic cell (DC) development, yet information about regulatory interactions between these signals remains limited. Here we show that the cytokines GM-CSF and Flt3L induce the transcriptional mediators Id2 and E2-2 and control DC lineage diversification by STAT-dependent pathways. We found that STAT5 is required for tissue CD103(+) DC generation and plasmacytoid DC (pDC) suppression in steady state or response to GM-CSF. STAT5 stimulates GM-CSF-dependent expression of Id2, which controls CD103(+) DC production and pDC inhibition. By contrast, pDCs, but not CD103(+) DCs, are dependent on STAT3. Consistently, STAT3 stimulates Flt3L-responsive expression of the pDC regulator Tcf4 (E2-2). These data suggest that STATs contribute to DC development by controlling transcription factors involved in lineage differentiation.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Differentiation/genetics , Dendritic Cells/physiology , Inhibitor of Differentiation Protein 2/genetics , STAT3 Transcription Factor/physiology , STAT5 Transcription Factor/physiology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Gene Expression Regulation, Developmental/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Inhibitor of Differentiation Protein 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Transcription Factor 4
SELECTION OF CITATIONS
SEARCH DETAIL