Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Immunity ; 56(5): 944-958.e6, 2023 05 09.
Article in English | MEDLINE | ID: mdl-37040761

ABSTRACT

Interferon-γ (IFN-γ) is a key cytokine in response to viral or intracellular bacterial infection in mammals. While a number of enhancers are described to promote IFN-γ responses, to the best of our knowledge, no silencers for the Ifng gene have been identified. By examining H3K4me1 histone modification in naive CD4+ T cells within Ifng locus, we identified a silencer (CNS-28) that restrains Ifng expression. Mechanistically, CNS-28 maintains Ifng silence by diminishing enhancer-promoter interactions within Ifng locus in a GATA3-dependent but T-bet-independent manner. Functionally, CNS-28 restrains Ifng transcription in NK cells, CD4+ cells, and CD8+ T cells during both innate and adaptive immune responses. Moreover, CNS-28 deficiency resulted in repressed type 2 responses due to elevated IFN-γ expression, shifting Th1 and Th2 paradigm. Thus, CNS-28 activity ensures immune cell quiescence by cooperating with other regulatory cis elements within the Ifng gene locus to minimize autoimmunity.


Subject(s)
CD8-Positive T-Lymphocytes , Interferon-gamma , Animals , Interferon-gamma/genetics , Interferon-gamma/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Regulatory Sequences, Nucleic Acid , Homeostasis , Th1 Cells , Mammals
2.
Immunity ; 55(4): 639-655.e7, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35381213

ABSTRACT

Adaptive CD4+ T helper cells and their innate counterparts, innate lymphoid cells, utilize an identical set of transcription factors (TFs) for their differentiation and functions. However, similarities and differences in the induction of these TFs in related lymphocytes are still elusive. Here, we show that T helper-1 (Th1) cells and natural killer (NK) cells displayed distinct epigenomes at the Tbx21 locus, which encodes T-bet, a critical TF for regulating type 1 immune responses. The initial induction of T-bet in NK precursors was dependent on the NK-specific DNase I hypersensitive site Tbx21-CNS-3, and the expression of the interleukin-18 (IL-18) receptor; IL-18 induced T-bet expression through the transcription factor RUNX3, which bound to Tbx21-CNS-3. By contrast, signal transducer and activator of transcription (STAT)-binding motifs within Tbx21-CNS-12 were critical for IL-12-induced T-bet expression during Th1 cell differentiation both in vitro and in vivo. Thus, type 1 innate and adaptive lymphocytes utilize distinct enhancer elements for their development and differentiation.


Subject(s)
Immunity, Innate , Interleukin-18 , Killer Cells, Natural , Th1 Cells , Cell Differentiation , Interleukin-18/metabolism , Killer Cells, Natural/immunology , T-Box Domain Proteins/metabolism , Th1 Cells/immunology , Transcription Factors/metabolism
3.
Immunity ; 53(4): 745-758.e4, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33010223

ABSTRACT

Innate immune responses rely on rapid and precise gene regulation mediated by accessibility of regulatory regions to transcription factors (TFs). In natural killer (NK) cells and other innate lymphoid cells, competent enhancers are primed during lineage acquisition, and formation of de novo enhancers characterizes the acquisition of innate memory in activated NK cells and macrophages. Here, we investigated how primed and de novo enhancers coordinate to facilitate high-magnitude gene induction during acute activation. Epigenomic and transcriptomic analyses of regions near highly induced genes (HIGs) in NK cells both in vitro and in a model of Toxoplasma gondii infection revealed de novo chromatin accessibility and enhancer remodeling controlled by signal-regulated TFs STATs. Acute NK cell activation redeployed the lineage-determining TF T-bet to de novo enhancers, independent of DNA-sequence-specific motif recognition. Thus, acute stimulation reshapes enhancer function through the combinatorial usage and repurposing of both lineage-determining and signal-regulated TFs to ensure an effective response.


Subject(s)
Enhancer Elements, Genetic/genetics , Enhancer Elements, Genetic/immunology , Killer Cells, Natural/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Animals , Chromatin/genetics , Chromatin/immunology , Female , Gene Expression/genetics , Gene Expression/immunology , Gene Expression Profiling/methods , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Immunity, Innate/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Toxoplasma/immunology , Toxoplasmosis/genetics , Toxoplasmosis/immunology
4.
Immunity ; 48(2): 227-242.e8, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29466755

ABSTRACT

How chromatin reorganization coordinates differentiation and lineage commitment from hematopoietic stem and progenitor cells (HSPCs) to mature immune cells has not been well understood. Here, we carried out an integrative analysis of chromatin accessibility, topologically associating domains, AB compartments, and gene expression from HSPCs to CD4+CD8+ T cells. We found that abrupt genome-wide changes at all three levels of chromatin organization occur during the transition from double-negative stage 2 (DN2) to DN3, accompanying the T lineage commitment. The transcription factor BCL11B, a critical regulator of T cell commitment, is associated with increased chromatin interaction, and Bcl11b deletion compromised chromatin interaction at its target genes. We propose that these large-scale and concerted changes in chromatin organization present an energy barrier to prevent the cell from reversing its fate to earlier stages or redirecting to alternatives and thus lock the cell fate into the T lineages.


Subject(s)
Cell Lineage , Cell Nucleus/physiology , Chromatin/physiology , T-Lymphocytes/physiology , Animals , Cell Differentiation , Humans , Repressor Proteins/physiology , Tumor Suppressor Proteins/physiology
5.
Proc Natl Acad Sci U S A ; 118(27)2021 07 06.
Article in English | MEDLINE | ID: mdl-34187897

ABSTRACT

Immunoglobulin A (IgA)-producing plasma cells derived from conventional B cells in the gut play an important role in maintaining the homeostasis of gut flora. Both T cell-dependent and T cell-independent IgA class switching occurs in the lymphoid structures in the gut, whose formation depends on lymphoid tissue inducers (LTis), a subset of innate lymphoid cells (ILCs). However, our knowledge on the functions of non-LTi helper-like ILCs, the innate counter parts of CD4 T helper cells, in promoting IgA production is still limited. By cell adoptive transfer and utilizing a unique mouse strain, we demonstrated that the generation of IgA-producing plasma cells from B cells in the gut occurred efficiently in the absence of both T cells and helper-like ILCs and without engaging TGF-ß signaling. Nevertheless, B cell recruitment and/or retention in the gut required functional NKp46-CCR6+ LTis. Therefore, while CCR6+ LTis contribute to the accumulation of B cells in the gut through inducing lymphoid structure formation, helper-like ILCs are not essential for the T cell-independent generation of IgA-producing plasma cells.


Subject(s)
B-Lymphocytes/immunology , Gastrointestinal Tract/immunology , Immunity, Innate , Immunoglobulin A/immunology , Immunoglobulin Class Switching , Lymphocytes/immunology , T-Lymphocytes/immunology , Animals , GATA3 Transcription Factor/metabolism , Immunoglobulin Class Switching/immunology , Integrases/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
6.
Cell Mol Life Sci ; 77(2): 289-303, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31432236

ABSTRACT

CD4 T-helper (Th) cells secret a variety of inflammatory cytokines and play critical roles in host defense against invading foreign pathogens. On the other hand, uncontrolled inflammatory responses mediated by Th cells may result in tissue damage and inflammatory disorders including autoimmune and allergic diseases. Thus, the induction of anti-inflammatory cytokine expression becomes an important "brake" to repress and/or terminate aberrant and/or unnecessary immune responses. Interleukin-10 (IL-10) is one of the most important anti-inflammatory cytokines to limit inflammatory Th cells and immunopathology and to maintain tissue homeostasis. Many studies have indicated that Th cells can be a major source of IL-10 under specific conditions both in mouse and human and that extracellular signals and cell intrinsic molecular switches are required to turn on and off Il10 expression in different Th cells. In this review, we will highlight the recent findings that have enhanced our understanding on the mechanisms of IL-10 induction in distinct Th-cell subsets, including Th1, Th2, and Th17 cells, as well as the importance of these IL-10-producing anti-inflammatory Th cells in immunity and inflammation.


Subject(s)
Anti-Inflammatory Agents/immunology , Cell Differentiation/immunology , Inflammation/immunology , Interleukin-10/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Humans
7.
bioRxiv ; 2024 May 20.
Article in English | MEDLINE | ID: mdl-38903096

ABSTRACT

The pair of transcription factors Bcl6-Blimp1 is well-known for follicular T helper (Tfh) cell fate determination, however, the mechanism(s) for Bcl6-independent regulation of CXCR5 during Tfh migration into germinal center (GC) is still unclear. In this study, we uncovered another pair of transcription factors, Bhlhe40-Pou2af1, that regulates CXCR5 expression. Pou2af1 was specifically expressed in Tfh cells whereas Bhlhe40 expression was found high in non-Tfh cells. Pou2af1 promoted Tfh formation and migration into GC by upregulating CXCR5 but not Bcl6, while Bhlhe40 repressed this process by inhibiting Pou2af1 expression. RNA-Seq analysis of antigen-specific Tfh cells generated in vivo confirmed the role of Bhlhe40-Pou2af1 axis in regulating optimal CXCR5 expression. Thus, the regulation of CXCR5 expression and migration of Tfh cells into GC involves a transcriptional regulatory circuit consisting of Bhlhe40 and Pou2af1, which operates independent of the Bcl6-Blimp1 circuit that determines the Tfh cell fate.

8.
J Biol Chem ; 287(1): 382-392, 2012 Jan 02.
Article in English | MEDLINE | ID: mdl-22084247

ABSTRACT

Janus kinase 1/signal transducers and activators of transcription 3 (JAK1/STAT3) pathway is one of the recognized oncogenic signaling pathways that frequently overactivated in a variety of human tumors. Despite rapid progress in elucidating the molecular mechanisms of activation of JAK/STAT pathway, the processes that regulate JAK/STAT deactivation need to be further clarified. Here we demonstrate that CUE domain-containing 2 (CUEDC2) inhibits cytokine-induced phosphorylation of JAK1 and STAT3 and the subsequent STAT3 transcriptional activity. Further analysis by a yeast two-hybrid assay showed that CUEDC2 could engage in a specific interaction with a key JAK/STAT inhibitor, SOCS3 (suppressors of cytokine signaling 3). The interaction between CUEDC2 and SOCS3 is required for the inhibitory effect of CUEDC2 on JAK1 and STAT3 activity. Additionally, we found CUEDC2 functions collaboratively with SOCS3 to inhibit JAK1/STAT3 signaling by increasing SOCS3 stability via enhancing its association with Elongin C. Therefore, our findings revealed a new biological activity for CUEDC2 as the regulator of JAK1/STAT3 signaling and paved the way to a better understanding of the mechanisms by which SOCS3 has been linked to suppression of the JAK/STAT pathway.


Subject(s)
Carrier Proteins/metabolism , Janus Kinase 1/metabolism , Membrane Proteins/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Suppressor of Cytokine Signaling Proteins/metabolism , Adaptor Proteins, Signal Transducing , Cell Line , Elongin , Enzyme Activation , Humans , Phosphorylation , Protein Stability , Proteolysis , Suppressor of Cytokine Signaling 3 Protein , Transcription Factors/metabolism , Transcription, Genetic
9.
Biochem Biophys Res Commun ; 431(4): 686-92, 2013 Feb 22.
Article in English | MEDLINE | ID: mdl-23357418

ABSTRACT

RIG-I/MDA5 plays a pivotal role in innate immunity by detecting intracellular double-stranded RNA (dsRNA) and activating the transcription of type I interferons and proinflammatory factors, but the exactly regulating mechanism of RIG-I/MDA5 signaling remains elusive. In this study, UbL-UBA domain containing protein RAD23A was identified as a negative regulator of RIG-I/MDA5-mediated signaling activation through a small interfering RNA (siRNA)-based screening. Knockdown of RAD23A augmented the expression of RIG-I/MDA5-mediated expression of proinflammatory cytokines and IFN-ß whereas ectopic expression of RAD23A showed the converse effect. Moreover, we confirmed the interaction between RAD23A and tumor necrosis factor receptor-associated factor 2 (TRAF2), an essential mediator of RIG-I/MDA5 signaling, and found that RAD23A down-regulated TRAF2 protein level through ubiquitin-proteasome system. Therefore, this study identified RAD23A as a novel negative regulator of RIG-I/MDA5 mediated anti-virus response.


Subject(s)
DEAD-box RNA Helicases/metabolism , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , TNF Receptor-Associated Factor 2/metabolism , Ubiquitination , DEAD Box Protein 58 , DNA Repair Enzymes/genetics , DNA, Viral/immunology , DNA-Binding Proteins/genetics , Gene Knockdown Techniques , HEK293 Cells , Humans , Interferon-Induced Helicase, IFIH1 , Interferon-beta/biosynthesis , NF-kappa B/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Structure, Tertiary , Proteolysis , RNA, Small Interfering/genetics , RNA, Viral/immunology , Receptors, Immunologic , Signal Transduction , Ubiquitin/metabolism
10.
Cell Rep ; 42(8): 112924, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37540600

ABSTRACT

Lymphoid tissue inducer (LTi) cells, a subset of innate lymphoid cells (ILCs), play an essential role in the formation of secondary lymphoid tissues. However, the regulation of the development and functions of this ILC subset is still elusive. In this study, we report that the transcription factor T cell factor 1 (TCF-1), just as GATA3, is indispensable for the development of non-LTi ILC subsets. While LTi cells are still present in TCF-1-deficient mice, the organogenesis of Peyer's patches (PPs), but not of lymph nodes, is impaired in these mice. LTi cells from different tissues have distinct gene expression patterns, and TCF-1 regulates the expression of lymphotoxin specifically in PP LTi cells. Mechanistically, TCF-1 may directly and/or indirectly regulate Lta, including through promoting the expression of GATA3. Thus, the TCF-1-GATA3 axis, which plays an important role during T cell development, also critically regulates the development of non-LTi cells and tissue-specific functions of LTi cells.


Subject(s)
Immunity, Innate , T Cell Transcription Factor 1 , Animals , Mice , Lymphocytes , Lymphoid Tissue/metabolism , T Cell Transcription Factor 1/metabolism
11.
Front Immunol ; 13: 1081153, 2022.
Article in English | MEDLINE | ID: mdl-36685550

ABSTRACT

CD4 T helper (Th) cell subsets, including Th1, Th2 and Th17 cells, and their innate counterparts innate lymphoid cell (ILC) subsets consisting of ILC1s, ILC2s and ILC3s, display similar effector cytokine-producing capabilities during pro-inflammatory immune responses. These lymphoid cell subsets utilize the same set of lineage-determining transcription factors (LDTFs) for their differentiation, development and functions. The distinct ontogeny and developmental niches between Th cells and ILCs indicate that they may adopt different external signals for the induction of LDTF during lineage commitment. Increasing evidence demonstrates that many conserved cis-regulatory elements at the gene loci of LDTFs are often preferentially utilized for the induction of LDTF expression during Th cell differentiation and ILC development at different stages. In this review, we discuss the functions of lineage-related cis-regulatory elements in inducing T-bet, GATA3 or RORγt expression based on the genetic evidence provided in recent publications. We also review and compare the upstream signals involved in LDTF induction in Th cells and ILCs both in vitro and in vivo. Finally, we discuss the possible mechanisms and physiological importance of regulating LDTF dynamic expression during ILC development and activation.


Subject(s)
Lymphocytes , Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Immunity, Innate , Gene Expression Regulation , T-Lymphocytes, Helper-Inducer
12.
Nat Commun ; 11(1): 4624, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32913264

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

13.
Nat Commun ; 11(1): 3366, 2020 07 06.
Article in English | MEDLINE | ID: mdl-32632165

ABSTRACT

CD4+ T lymphocytes consist of naïve, antigen-specific memory, and memory-phenotype (MP) cell compartments at homeostasis. We recently showed that MP cells exert innate-like effector function during host defense, but whether MP CD4+ T cells are functionally heterogeneous and, if so, what signals specify the differentiation of MP cell subpopulations under homeostatic conditions is still unclear. Here we characterize MP lymphocytes as consisting of T-bethigh, T-betlow, and T-bet- subsets, with innate, Th1-like effector activity exclusively associated with T-bethigh cells. We further show that the latter population depends on IL-12 produced by CD8α+ type 1 dendritic cells (DC1) for its differentiation. Finally, our data demonstrate that this tonic IL-12 production requires TLR-MyD88 signaling independent of foreign agonists, and is further enhanced by CD40-CD40L interactions between DC1 and CD4+ T lymphocytes. We propose that optimal differentiation of T-bethigh MP lymphocytes at homeostasis is driven by self-recognition signals at both the DC and Tcell levels.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Homeostasis/immunology , Immunologic Memory/immunology , T-Box Domain Proteins/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD40 Antigens/immunology , CD40 Antigens/metabolism , CD40 Ligand/genetics , CD40 Ligand/immunology , CD40 Ligand/metabolism , CD8 Antigens/immunology , CD8 Antigens/metabolism , Cell Communication/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Interleukin-12/genetics , Interleukin-12/immunology , Interleukin-12/metabolism , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Signal Transduction/immunology , T-Box Domain Proteins/metabolism , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/metabolism
14.
J Exp Med ; 215(11): 2705-2714, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30232200

ABSTRACT

T follicular helper (Tfh) cells express transcription factor BCL-6 and cytokine IL-21. Mature Tfh cells are also capable of producing IFN-γ without expressing the Th1 transcription factor T-bet. Whether this IFN-γ-producing Tfh population represents a unique Tfh subset with a distinct differentiation pathway is poorly understood. By using T-bet fate-mapping mouse strains, we discovered that almost all the IFN-γ-producing Tfh cells have previously expressed T-bet and express high levels of NKG2D. DNase I hypersensitivity analysis indicated that the Ifng gene locus is partially accessible in this "ex-T-bet" population with a history of T-bet expression. Furthermore, multicolor tissue imaging revealed that the ex-T-bet Tfh cells found in germinal centers express IFN-γ in situ. Finally, we found that IFN-γ-expressing Tfh cells are absent in T-bet-deficient mice, but fully present in mice with T-bet deletion at late stages of T cell differentiation. Together, our findings demonstrate that transient expression of T-bet epigenetically imprints the Ifng locus for cytokine production in this Th1-like Tfh cell subset.


Subject(s)
Cell Differentiation/immunology , Genomic Imprinting/immunology , Germinal Center/immunology , T-Box Domain Proteins/immunology , Th1 Cells/immunology , Animals , Cell Differentiation/genetics , Germinal Center/cytology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukins/genetics , Interleukins/immunology , Male , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/immunology , T-Box Domain Proteins/genetics , Th1 Cells/cytology
15.
J Exp Med ; 215(5): 1449-1462, 2018 05 07.
Article in English | MEDLINE | ID: mdl-29514917

ABSTRACT

GATA-binding protein 3 (GATA3) acts as the master transcription factor for type 2 T helper (Th2) cell differentiation and function. However, it is still elusive how GATA3 function is precisely regulated in Th2 cells. Here, we show that the transcription factor B cell lymphoma 11b (Bcl11b), a previously unknown component of GATA3 transcriptional complex, is involved in GATA3-mediated gene regulation. Bcl11b binds to GATA3 through protein-protein interaction, and they colocalize at many important cis-regulatory elements in Th2 cells. The expression of type 2 cytokines, including IL-4, IL-5, and IL-13, is up-regulated in Bcl11b-deficient Th2 cells both in vitro and in vivo; such up-regulation is completely GATA3 dependent. Genome-wide analyses of Bcl11b- and GATA3-regulated genes (from RNA sequencing), cobinding patterns (from chromatin immunoprecipitation sequencing), and Bcl11b-modulated epigenetic modification and gene accessibility suggest that GATA3/Bcl11b complex is involved in limiting Th2 gene expression, as well as in inhibiting non-Th2 gene expression. Thus, Bcl11b controls both GATA3-mediated gene activation and repression in Th2 cells.


Subject(s)
Cell Differentiation , GATA3 Transcription Factor/metabolism , Repressor Proteins/metabolism , Th1 Cells/cytology , Th2 Cells/cytology , Tumor Suppressor Proteins/metabolism , Animals , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Cell Proliferation , Chromatin/metabolism , Cytokines/metabolism , Epigenesis, Genetic , Genome , Lectins, C-Type/metabolism , Mice, Inbred C57BL , Protein Binding
16.
J Exp Med ; 214(7): 1861-1876, 2017 Jul 03.
Article in English | MEDLINE | ID: mdl-28630089

ABSTRACT

CD4 T cells, including T regulatory cells (Treg cells) and effector T helper cells (Th cells), and recently identified innate lymphoid cells (ILCs) play important roles in host defense and inflammation. Both CD4 T cells and ILCs can be classified into distinct lineages based on their functions and the expression of lineage-specific genes, including those encoding effector cytokines, cell surface markers, and key transcription factors. It was first recognized that each lineage expresses a specific master transcription factor and the expression of these factors is mutually exclusive because of cross-regulation among these factors. However, recent studies indicate that the master regulators are often coexpressed. Furthermore, the expression of master regulators can be dynamic and quantitative. In this review, we will first discuss similarities and differences between the development and functions of CD4 T cell and ILC subsets and then summarize recent literature on quantitative, dynamic, and cell type-specific balance between the master transcription factors in determining heterogeneity and plasticity of these subsets.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunity, Innate/immunology , Transcription Factors/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Lineage/genetics , Cell Lineage/immunology , Gene Expression/immunology , Humans , Immunity, Innate/genetics , Models, Immunological , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
17.
Nat Commun ; 7: 11378, 2016 04 25.
Article in English | MEDLINE | ID: mdl-27109496

ABSTRACT

The intimate link between location of fat accumulation and metabolic disease risk and depot-specific differences is well established, but how these differences between depots are regulated at the molecular level remains largely unclear. Here we show that TRIP-Br2 mediates endoplasmic reticulum (ER) stress-induced inflammatory responses in visceral fat. Using in vitro, ex vivo and in vivo approaches, we demonstrate that obesity-induced circulating factors upregulate TRIP-Br2 specifically in visceral fat via the ER stress pathway. We find that ablation of TRIP-Br2 ameliorates both chemical and physiological ER stress-induced inflammatory and acute phase response in adipocytes, leading to lower circulating levels of inflammatory cytokines. Using promoter assays, as well as molecular and pharmacological experiments, we show that the transcription factor GATA3 is responsible for the ER stress-induced TRIP-Br2 expression in visceral fat. Taken together, our study identifies molecular regulators of inflammatory response in visceral fat that-given that these pathways are conserved in humans-might serve as potential therapeutic targets in obesity.


Subject(s)
Endoplasmic Reticulum Stress , GATA3 Transcription Factor/immunology , Intra-Abdominal Fat/immunology , Obesity/immunology , Transcription Factors/immunology , Adipocytes/immunology , Adipokines/genetics , Adipokines/immunology , Animals , GATA3 Transcription Factor/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Obesity/genetics , Obesity/physiopathology , Transcription Factors/genetics
18.
Nat Commun ; 5: 4513, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-25072696

ABSTRACT

CD40, a member of tumour necrosis factor receptor (TNFR) superfamily, has a pivotal role in B-cell-mediated immunity through various effector pathways including AKT kinase, but the signal transduction of CD40-meidated AKT activation is poorly understood. Here we report that the neural precursor cell expressed developmentally downregulated protein 4 (NEDD4), homologous to E6-AP Carboxyl Terminus family E3 ubiquitin ligase, is a novel component of the CD40 signalling complex. It has a key role in CD40-mediated AKT activation and is involved in modulating immunoglobulin class switch through regulating the expression of activation-induced cytidine deaminase. NEDD4 constitutively interacts with CD40 and mediates K63-linked ubiquitination of TNFR-associated factor3 (TRAF3). The ubiquitination of TRAF3 by NEDD4 is critical for CD40-mediated AKT activation. Thus, NEDD4 is a previously unknown component of the CD40 signalling complex necessary for AKT activation.


Subject(s)
CD40 Antigens/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Immunity, Cellular/immunology , Oncogene Protein v-akt/metabolism , Signal Transduction/immunology , TNF Receptor-Associated Factor 3/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Flow Cytometry , Humans , Immunoprecipitation , Mass Spectrometry , Mice , Mice, Knockout , Nedd4 Ubiquitin Protein Ligases , RNA Interference , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Ubiquitination
19.
Cell Rep ; 7(6): 1982-93, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24882011

ABSTRACT

CUEDC2, a CUE-domain-containing protein, modulates inflammation, but its involvement in tumorigenesis is still poorly understood. Here, we report that CUEDC2 is a key regulator of macrophage function and critical for protection against colitis-associated tumorigenesis. CUEDC2 expression is dramatically upregulated during macrophage differentiation, and CUEDC2 deficiency results in excessive production of proinflammatory cytokines. The level of CUEDC2 in macrophages is modulated by miR- 324-5p. We find that Cuedc2 KO mice are more susceptible to dextran-sodium-sulfate-induced colitis, and macrophage transplantation results suggest that the increased susceptibility results from the dysfunction of macrophages lacking CUEDC2. Furthermore, we find that Cuedc2 KO mice are more prone to colitis-associated cancer. Importantly, CUEDC2 expression is almost undetectable in macrophages in human colon cancer, and this decreased CUEDC2 expression is associated with high levels of interleukin-4 and miR-324-5p. Thus, CUEDC2 plays a crucial role in modulating macrophage function and is associated with both colitis and colon tumorigenesis.


Subject(s)
Carrier Proteins/metabolism , Colonic Neoplasms/metabolism , Macrophages/metabolism , Membrane Proteins/metabolism , MicroRNAs/metabolism , Repressor Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Carrier Proteins/immunology , Colitis/genetics , Colitis/immunology , Colitis/metabolism , Colitis/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Female , Gene Expression Regulation , HeLa Cells , Humans , Macrophage Activation , Macrophages/immunology , Macrophages/pathology , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Transgenic , MicroRNAs/genetics , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Repressor Proteins/immunology , Signal Transduction
20.
FEBS Lett ; 587(6): 607-13, 2013 Mar 18.
Article in English | MEDLINE | ID: mdl-23416296

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) is a critical transcriptional factor in a variety of cellular processes, and is frequently over-activated in a range of human tumors. However, the processes that regulate STAT3 activation need to be further clarified. With a yeast two-hybrid screening, we identified enoyl-CoA hydratase short chain 1 (ECHS1) as a novel STAT3 binding protein. We further confirmed the interaction between STAT3 and ECHS1 by GST-pull down and co-immnunoprecipitation. Importantly, we found that ECHS1 specifically represses STAT3 activity and negatively regulates the expression of several target genes of STAT3 through inhibiting STAT3 phosphorylation. Therefore, our findings will provide new insights into the mechanism of STAT3 signaling regulation.


Subject(s)
Enoyl-CoA Hydratase/genetics , Gene Expression Regulation, Neoplastic , STAT3 Transcription Factor/genetics , Enoyl-CoA Hydratase/metabolism , Epidermal Growth Factor/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , HeLa Cells , Humans , Immunoprecipitation , Interleukin-6/pharmacology , Isoenzymes/genetics , Isoenzymes/metabolism , Phosphorylation , Protein Binding , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL