Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
iScience ; 26(8): 107474, 2023 Aug 18.
Article in English | MEDLINE | ID: mdl-37583551

ABSTRACT

Targeted deletion of TRAF7 revealed that it is a crucial part of shear stress-responsive MEKK3-MEK5-ERK5 signaling pathway induced in endothelial cells by blood flow. Similar to Mekk3-, Mek5- or Erk5-deficient mice, Traf7-deficient embryos died in utero around midgestation due to impaired endothelium integrity. They displayed significantly lower expression of transcription factor Klf2, an essential regulator of vascular hemodynamic forces downstream of the MEKK3-MEK-ERK5 signaling pathway. In addition, deletion of Traf7 in endothelial cells of postnatal mice was associated with severe cerebral hemorrhage. Here, we show that besides MEKK3 and MEK5, TRAF7 associates with a planar cell polarity protein SCRIB. SCRIB binds with an N-terminal region of TRAF7, while MEKK3 associates with the C-terminal WD40 domain. Downregulation of TRAF7 as well as SCRIB inhibited fluid shear stress-induced phosphorylation of ERK5 in cultured endothelial cells. These findings suggest that TRAF7 and SCRIB may comprise an upstream part of the MEKK3-MEK5-ERK5 signaling pathway.

2.
Front Oncol ; 13: 1126550, 2023.
Article in English | MEDLINE | ID: mdl-36937440

ABSTRACT

Introduction: Meningiomas are the most common primary central nervous system (CNS) tumors in adults, representing approximately one-third of all primary adult CNS tumors. Although several recent publications have proposed alternative grading systems of meningiomas that incorporate genomic and/or epigenomic data to better predict meningioma recurrence and progression-free survival, our understanding of driving forces of meningioma development is still limited. Objective: To define gene expression signatures of the most common subtypes of meningiomas to better understand cellular processes and signaling pathways specific for each tumor genotype. Methods: We used RNA sequencing (RNA-seq) to determine whole transcriptome profiles of twenty meningiomas with genomic alterations including NF2 inactivation, loss of chr1p, and missense mutations in TRAF7, AKT1 and KLF4. Results: The analysis revealed that meningiomas with NF2 gene inactivation expressed higher levels of BCL2 and GLI1 compared with tumors harboring TRAF7 missense mutations. Moreover, NF2 meningiomas were subdivided into two distinct groups based on additional loss of chr1p. NF2 tumors with intact chr1p were characterized by the high expression of tumor suppressor PTCH2 compared to NF2 tumors with chr1p loss. Taken together with the high expression of BCL2 and GLI1, these results suggest that activation of Sonic Hedgehog pathway may contribute to NF2 meningioma development. In contrast, NF2 tumors with chr1p loss expressed high levels of transcription factor FOXD3 and its antisense RNA FOXD3-AS1. Examination of TRAF7 tumors demonstrated that TRAF7 regulates a number of biomechanically responsive genes (KRT6a, KRT16, IL1RL1, and AQP3 among others). Interestingly, AKT1 and KLF4 meningiomas expressed genes specific for PI3K/AKT signaling pathway, suggesting overlapping gene signatures between the two subtypes. In addition, KLF4 meningiomas had high expression of carcinoembryonic antigen family members CEACAM6 and CEACAM5. Conclusions: Each group of meningiomas displayed a unique gene expression signature suggesting signaling pathways potentially implicated in tumorigenesis. These findings will improve our understanding of meningioma tumorigenesis and prognosis.

3.
J Dev Biol ; 11(1)2023 Mar 16.
Article in English | MEDLINE | ID: mdl-36976102

ABSTRACT

The COMMD (copper metabolism MURR1 domain containing) family includes ten structurally conserved proteins (COMMD1 to COMMD10) in eukaryotic multicellular organisms that are involved in a diverse array of cellular and physiological processes, including endosomal trafficking, copper homeostasis, and cholesterol metabolism, among others. To understand the role of COMMD10 in embryonic development, we used Commd10Tg(Vav1-icre)A2Kio/J mice, where the Vav1-cre transgene is integrated into an intron of the Commd10 gene, creating a functional knockout of Commd10 in homozygous mice. Breeding heterozygous mice produced no COMMD10-deficient (Commd10Null) offspring, suggesting that COMMD10 is required for embryogenesis. Analysis of Commd10Null embryos demonstrated that they displayed stalled development by embryonic day 8.5 (E8.5). Transcriptome analysis revealed that numerous neural crest-specific gene markers had lower expression in mutant versus wild-type (WT) embryos. Specifically, Commd10Null embryos displayed significantly lower expression levels of a number of transcription factors, including a major regulator of the neural crest, Sox10. Moreover, several cytokines/growth factors involved in early embryonic neurogenesis were also lower in mutant embryos. On the other hand, Commd10Null embryos demonstrated higher expression of genes involved in tissue remodeling and regression processes. Taken together, our findings show that Commd10Null embryos die by day E8.5 due to COMMD10-dependent neural crest failure, revealing a new and critical role for COMMD10 in neural development.

4.
iScience ; 25(8): 104839, 2022 Aug 19.
Article in English | MEDLINE | ID: mdl-35996584

ABSTRACT

Krüppel-like factor 4 (KLF4) is a transcription factor that has been proven necessary for both induction and maintenance of pluripotency and self-renewal. Whole-genome sequencing defined a unique mutation in KLF4 (KLF4K409Q) in human meningiomas. However, the molecular mechanism of this tumor-specific KLF4 mutation is unknown. Using genome-wide high-throughput and focused quantitative transcriptional approaches in human cell lines, primary meningeal cells, and meningioma tumor tissue, we found that a change in the evolutionarily conserved DNA-binding domain of KLF4 alters its DNA recognition preference, resulting in a shift in downstream transcriptional activity. In the KLF4K409Q-specific targets, the normally silent fibroblast growth factor 3 (FGF3) is activated. We demonstrated a neomorphic function of KLF4K409Q in stimulating FGF3 transcription through binding to its promoter and in using short tandem repeats (STRs) located within the locus as enhancers.

5.
J Immunol ; 205(9): 2479-2488, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32978279

ABSTRACT

The human TNF/LT locus genes TNF, LTA, and LTB are expressed in a cell type-specific manner. In this study, we show that a highly conserved NFAT binding site within the distal noncoding element hHS-8 coordinately controls TNF and LTA gene expression in human T cells. Upon activation of primary human CD4+ T cells, hHS-8 and the TNF and LTA promoters display increased H3K27 acetylation and nuclease sensitivity and coordinate induction of TNF, LTA, and hHS-8 enhancer RNA transcription occurs. Functional analyses using CRISPR/dead(d)Cas9 targeting of the hHS-8-NFAT site in the human T cell line CEM demonstrate significant reduction of TNF and LTA mRNA synthesis and of RNA polymerase II recruitment to their promoters. These studies elucidate how a distal element regulates the inducible cell type-specific gene expression program of the human TNF/LT locus and provide an approach for modulation of TNF and LTA transcription in human disease using CRISPR/dCas9.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Gene Expression/genetics , Lymphotoxin-alpha/genetics , Tumor Necrosis Factor-alpha/genetics , Acetylation , Binding Sites/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Conserved Sequence/genetics , Enhancer Elements, Genetic/genetics , Histones/genetics , Humans , Leukocytes, Mononuclear/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/genetics , RNA Polymerase II/genetics , RNA, Messenger/genetics , THP-1 Cells/metabolism , Transcription, Genetic/genetics
6.
PLoS One ; 7(7): e41427, 2012.
Article in English | MEDLINE | ID: mdl-22844476

ABSTRACT

In T cells, the transcription factor nuclear factor of activated T cells p (NFATp) is a key regulator of the cytokine genes tumor necrosis factor (TNF) and interferon-γ (IFN-γ). Here, we show that NFATp-deficient (NFATp(-/-)) mice have a dramatic and highly significant increase in mortality after Mycobacterium tuberculosis (MTb) infection as compared to mortality of control animals after MTb infection. Animals deficient in NFATp have significantly impaired levels of TNF and IFN-γ transcription and protein expression in naïve or total CD4(+) T cells, but display wild-type levels of TNF mRNA or protein from MTb-stimulated dendritic cells (DC). The rapid mortality and disease severity observed in MTb-infected NFATp(-/-) mice is associated with dysregulated production of TNF and IFN-γ in the lungs, as well as with increased levels of TNF, in their serum. Furthermore, global blocking of TNF production by injection of a TNF neutralizaing agent at 6 weeks, but not 12 weeks, post-MTb-infection further decreased the survival rate of both wild-type and NFATp(-/-) mice, indicating an early role for TNF derived from cells from the monocyte lineage in containment of infection. These results thus demonstrate that NFATp plays a critical role in immune containment of TB disease in vivo, through the NFATp-dependent expression of TNF and IFN-γ in T cells.


Subject(s)
NFATC Transcription Factors/metabolism , Tuberculosis, Pulmonary/metabolism , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Disease Susceptibility , Gene Deletion , Interferon-gamma/genetics , Lung/immunology , Lung/metabolism , Lung/microbiology , Mice , Monocytes/immunology , Monocytes/metabolism , Monocytes/microbiology , Mycobacterium tuberculosis/pathogenicity , NFATC Transcription Factors/deficiency , NFATC Transcription Factors/genetics , Signal Transduction , Tuberculosis, Pulmonary/genetics , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/pathology , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
7.
J Biol Chem ; 286(51): 44126-44133, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22027829

ABSTRACT

Regulation of TNF gene expression is cell type- and stimulus-specific. We have previously identified highly conserved noncoding regulatory elements within DNase I-hypersensitive sites (HSS) located 9 kb upstream (HSS-9) and 3 kb downstream (HSS+3) of the TNF gene, which play an important role in the transcriptional regulation of TNF in T cells. They act as enhancers and interact with the TNF promoter and with each other, generating a higher order chromatin structure. Here, we report a novel monocyte-specific AT-rich DNase I-hypersensitive element located 7 kb upstream of the TNF gene (HSS-7), which serves as a matrix attachment region in monocytes. We show that HSS-7 associates with topoisomerase IIα (Top2) in vivo and that induction of endogenous TNF mRNA expression is suppressed by etoposide, a Top2 inhibitor. Moreover, Top2 binds to and cleaves HSS-7 in in vitro analysis. Thus, HSS-7, which is selectively accessible in monocytes, can tether the TNF locus to the nuclear matrix via matrix attachment region formation, potentially promoting TNF gene expression by acting as a Top2 substrate.


Subject(s)
Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Gene Expression Regulation , Monocytes/cytology , Base Sequence , Cell Nucleus/metabolism , Chromatin/metabolism , DNA/metabolism , Deoxyribonuclease I/metabolism , HeLa Cells , Histones , Humans , Molecular Sequence Data , Poly-ADP-Ribose Binding Proteins , T-Lymphocytes/cytology , Transcription, Genetic
8.
Curr Dir Autoimmun ; 11: 27-60, 2010.
Article in English | MEDLINE | ID: mdl-20173386

ABSTRACT

The cytokine TNF is a critical mediator of immune and inflammatory responses. The TNF gene is an immediate early gene, rapidly transcribed in a variety of cell types following exposure to a broad range of pathogens and signals of inflammation and stress. Regulation of TNF gene expression at the transcriptional level is cell type- and stimulus-specific, involving the recruitment of distinct sets of transcription factors to a compact and modular promoter region. In this review, we describe our current understanding of the mechanisms through which TNF transcription is specifically activated by a variety of extracellular stimuli in multiple cell types, including T cells, B cells, macrophages, mast cells, dendritic cells, and fibroblasts. We discuss the role of nuclear factor of activated T cells and other transcription factors and coactivators in enhanceosome formation, as well as the contradictory evidence for a role for nuclear factor kappaB as a classical activator of the TNF gene. We describe the impact of evolutionarily conserved cis-regulatory DNA motifs in the TNF locus upon TNF gene transcription, in contrast to the neutral effect of single nucleotide polymorphisms. We also assess the regulatory role of chromatin organization, epigenetic modifications, and long-range chromosomal interactions at the TNF locus.


Subject(s)
Tumor Necrosis Factor-alpha/genetics , Animals , Base Sequence , Chromatin Assembly and Disassembly , DNA/genetics , Epigenesis, Genetic , Genes, Immediate-Early , Humans , Mice , Molecular Sequence Data , NF-kappa B/immunology , NFATC Transcription Factors/immunology , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Transcription Factors/immunology , Transcription, Genetic , Transcriptional Activation , Tumor Necrosis Factor-alpha/physiology
9.
Proc Natl Acad Sci U S A ; 105(50): 19637-42, 2008 Dec 16.
Article in English | MEDLINE | ID: mdl-19060202

ABSTRACT

The transcription factor NFATp integrates multiple signal transduction pathways through coordinate binding with basic-region leucine zipper (bZIP) proteins and other transcription factors. The NFATp monomer, even in the absence of its activation domains, recruits bZIP proteins to canonical NFAT-bZIP composite DNA elements. By contrast, the NFATp dimer and its bZIP partner bind noncooperatively to the NFAT-bZIP element of the tumor necrosis factor (TNF) gene promoter. This observation raises the possibility that the function of the activation domains of NFATp is dimer-specific. Here, we determine the consensus DNA binding site of the NFATp dimer, describe monomer- and dimer-specific NFATp-DNA contact patterns, and demonstrate that NFATp dimerization and dimer-specific activation subdomains are required for transcriptional activation from the TNF NFAT-bZIP element. We also show that these NFATp subdomains interact with the coactivator CBP (CREB-binding protein), which is required for NFATp-dependent TNF gene transcription. Thus, the context-specific function of the activation domains of NFAT can be potentiated by DNA-directed dimerization.


Subject(s)
NFATC Transcription Factors/metabolism , Base Sequence , Basic-Leucine Zipper Transcription Factors/metabolism , CREB-Binding Protein/metabolism , Consensus Sequence , DNA/metabolism , Dimerization , Electrophoretic Mobility Shift Assay , NFATC Transcription Factors/chemistry , NFATC Transcription Factors/genetics , Promoter Regions, Genetic , Protein Structure, Tertiary , Transcription, Genetic , Tumor Necrosis Factor-alpha/genetics
10.
Proc Natl Acad Sci U S A ; 104(43): 16850-5, 2007 Oct 23.
Article in English | MEDLINE | ID: mdl-17940009

ABSTRACT

Here we provide a mechanism for specific, efficient transcription of the TNF gene and, potentially, other genes residing within multigene loci. We identify and characterize highly conserved noncoding elements flanking the TNF gene, which undergo activation-dependent intrachromosomal interactions. These elements, hypersensitive site (HSS)-9 and HSS+3 (9 kb upstream and 3 kb downstream of the TNF gene, respectively), contain DNase I hypersensitive sites in naive, T helper 1, and T helper 2 primary T cells. Both HSS-9 and HSS+3 inducibly associate with acetylated histones, indicative of chromatin remodeling, bind the transcription factor nuclear factor of activated T cells (NFAT)p in vitro and in vivo, and function as enhancers of NFAT-dependent transactivation mediated by the TNF promoter. Using the chromosome conformation capture assay, we demonstrate that upon T cell activation intrachromosomal looping occurs in the TNF locus. HSS-9 and HSS+3 each associate with the TNF promoter and with each other, circularizing the TNF gene and bringing NFAT-containing nucleoprotein complexes into close proximity. TNF gene regulation thus reveals a mode of intrachromosomal interaction that combines a looped gene topology with interactions between enhancers and a gene promoter.


Subject(s)
Chromosomes, Mammalian/metabolism , Enhancer Elements, Genetic/genetics , Promoter Regions, Genetic/genetics , Tumor Necrosis Factor-alpha/genetics , Acetylation , Animals , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Deoxyribonuclease I/metabolism , Histones/metabolism , Lymphotoxin-alpha/genetics , Mice , Models, Genetic , NFATC Transcription Factors/metabolism , Protein Binding , Transcription Factor RelA/metabolism , Transcription, Genetic
11.
J Biol Chem ; 282(16): 11629-38, 2007 Apr 20.
Article in English | MEDLINE | ID: mdl-17303559

ABSTRACT

The tumor necrosis factor (TNF) gene is activated by multiple extracellular signals in a stimulus- and cell type-specific fashion. Based on the presence of kappaB-like DNA motifs in the region upstream of the TNF gene, some have proposed a direct role for NF-kappaB in lipopolysaccharide (LPS)-induced TNF gene transcription in cells of the monocyte/macrophage lineage. However, we have previously demonstrated a general and critical role for a minimal TNF promoter region bearing only one of the kappaB-like motifs, kappa3, which is bound by nuclear factor of activated T cell proteins in lymphocytes and fibroblasts in response to multiple stimuli and Ets proteins in LPS-stimulated macrophages. Here, in an effort to resolve these contrasting findings, we used a combination of site-directed mutagenesis of the TNF promoter, quantitative DNase I footprinting, and analysis of endogenous TNF mRNA production in response to multiple stimuli under conditions that inhibit NF-kappaB activation (using the proteasome inhibitor lactacystin and using cells lacking either functional NF-kappaB essential modulator, which is the IkappaB kinase regulatory subunit, or the Nemo gene itself). We find that TNF mRNA production in response to ionophore is NF-kappaB-independent, but inhibition of NF-kappaB activation attenuates virus- and LPS-induced TNF mRNA levels after initial induction. We conclude that induction of TNF gene transcription by virus or LPS does not depend upon NF-kappaB binding to the proximal promoter; rather, a stimulus-specific post-induction mechanism involving NF-kappaB, yet to be characterized, is involved in the maintenance of maximal TNF mRNA levels.


Subject(s)
Gene Expression Regulation , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/metabolism , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Amino Acid Motifs , Animals , DNA/chemistry , Deoxyribonuclease I/metabolism , Humans , Ionophores/pharmacology , Lipopolysaccharides/metabolism , Mice , NF-kappa B/metabolism , Proteasome Inhibitors , Protein Structure, Tertiary
12.
PLoS Pathog ; 2(12): e130, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17173480

ABSTRACT

To replicate, HIV-1 capitalizes on endogenous cellular activation pathways resulting in recruitment of key host transcription factors to its viral enhancer. RNA interference has been a powerful tool for blocking key checkpoints in HIV-1 entry into cells. Here we apply RNA interference to HIV-1 transcription in primary macrophages, a major reservoir of the virus, and specifically target the transcription factor NFAT5 (nuclear factor of activated T cells 5), which is the most evolutionarily divergent NFAT protein. By molecularly cloning and sequencing isolates from multiple viral subtypes, and performing DNase I footprinting, electrophoretic mobility shift, and promoter mutagenesis transfection assays, we demonstrate that NFAT5 functionally interacts with a specific enhancer binding site conserved in HIV-1, HIV-2, and multiple simian immunodeficiency viruses. Using small interfering RNA to ablate expression of endogenous NFAT5 protein, we show that the replication of three major HIV-1 viral subtypes (B, C, and E) is dependent upon NFAT5 in human primary differentiated macrophages. Our results define a novel host factor-viral enhancer interaction that reveals a new regulatory role for NFAT5 and defines a functional DNA motif conserved across HIV-1 subtypes and representative simian immunodeficiency viruses. Inhibition of the NFAT5-LTR interaction may thus present a novel therapeutic target to suppress HIV-1 replication and progression of AIDS.


Subject(s)
HIV-1/physiology , Macrophages/virology , RNA Interference , Terminal Repeat Sequences/physiology , Transcription Factors/physiology , Virus Replication/physiology , Acquired Immunodeficiency Syndrome/physiopathology , Cell Line , Cells, Cultured , DNA, Viral , Disease Progression , Gene Expression Regulation , HIV-1/genetics , HIV-1/pathogenicity , HeLa Cells , Humans , Macrophages/physiology , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Terminal Repeat Sequences/genetics , Transcription Factors/genetics , Virus Replication/genetics
13.
Nucleic Acids Res ; 33(12): 3845-54, 2005.
Article in English | MEDLINE | ID: mdl-16027109

ABSTRACT

Tumor necrosis factor (TNF) is a pro-inflammatory cytokine that plays an important role in a variety of infectious and autoimmune disorders. Its transcription is regulated in a stimulus- and cell-type-specific manner via the recruitment of distinct DNA/activator complexes forming secondary structures or enhanceosomes. NFATp, a member of the nuclear factor of activated T cells (NFAT) family of transcription factors, plays a critical role in TNF gene regulation under a variety of conditions. In this study, we show that NFAT5, the most recently described NFAT family member, binds to the TNF promoter in a manner distinct from other NFAT proteins and is a key mediator in the activation of TNF gene transcription during hypertonic stress alone.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Transcription Factors/metabolism , Transcriptional Activation , Tumor Necrosis Factor-alpha/genetics , Animals , Binding Sites , Cell Line , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Genes, Reporter , Mice , Mutation , NFATC Transcription Factors , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Osmotic Pressure , RNA Interference , RNA, Messenger/biosynthesis , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/biosynthesis
14.
J Immunol ; 175(2): 1041-6, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-16002704

ABSTRACT

The molecular mechanisms that regulate expression of the immunosuppressive cytokine IL-10 remain poorly understood. In this study, by measuring sensitivity to DNase I digestion, we show that production of IL-10 by primary mouse bone marrow-derived macrophages stimulated through pattern recognition receptors was associated with chromatin remodeling of the IL-10 locus. We also demonstrate that the IL-10 locus is remodeled in primary Th2 cells and IL-10-producing regulatory T cells that have been differentiated in vitro. Strikingly, a novel DNase I-hypersensitive site (HSS-4.5) was identified in stimulated macrophages, but not in T cells. We show that hyperacetylated histones were recruited to this site in stimulated macrophages. Furthermore, HSS-4.5 is highly conserved and contains a putative NF-kappaB binding site. In support of a function for this site, NF-kappaB p65/RelA was recruited to HSS-4.5 in vivo and its activation was required for optimal IL-10 gene expression in LPS-stimulated macrophages.


Subject(s)
Chromatin/immunology , Chromatin/metabolism , Interleukin-10/biosynthesis , Interleukin-10/genetics , Macrophages/immunology , Macrophages/metabolism , Animals , Base Sequence , Bone Marrow Cells/enzymology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Chromatin/genetics , Dendritic Cells/enzymology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Deoxyribonuclease I/genetics , Deoxyribonuclease I/metabolism , Genetic Markers/immunology , Humans , Interleukin-10/metabolism , Lipopolysaccharides/pharmacology , Macrophage Activation/genetics , Macrophage Activation/immunology , Macrophages/enzymology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , NF-kappa B/biosynthesis , NF-kappa B/metabolism , Protein Transport/genetics , Protein Transport/immunology , Regulatory Sequences, Nucleic Acid/immunology , Resting Phase, Cell Cycle/genetics , Resting Phase, Cell Cycle/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th2 Cells/cytology , Th2 Cells/immunology , Th2 Cells/metabolism , Transcription Factor RelA
15.
Mol Cell Biol ; 23(2): 526-33, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12509451

ABSTRACT

Tumor necrosis factor alpha (TNF-alpha) plays an important role in host containment of infection by Mycobacterium tuberculosis, one of the leading causes of death by an infectious agent globally. Using the pathogenic M. tuberculosis strain H37Rv, we present evidence that upon stimulation of monocytic cells by M. tuberculosis a unique TNF-alpha enhanceosome is formed, and it is distinct from the TNF-alpha enhanceosome that forms in T cells stimulated by antigen engagement or virus infection. A distinct set of activators including ATF-2, c-jun, Ets, Sp1, Egr-1 and the coactivator proteins CBP/p300 are recruited to the TNF-alpha promoter after stimulation with M. tuberculosis. Furthermore, the formation of this enhanceosome is dependent on inducer-specific helical phasing relationships between transcription factor binding sites. We also show that the transcriptional activity of CBP/p300 is potentiated by mycobacterial stimulation of monocytes. The identification of TNF-alpha regulatory elements and coactivators involved in M. tuberculosis-stimulated gene expression thus provides potential selective molecular targets in the modulation of TNF-alpha gene expression in the setting of mycobacterial infection.


Subject(s)
Gene Expression Regulation , Immediate-Early Proteins , Mycobacterium tuberculosis/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Activating Transcription Factor 2 , Animals , Base Sequence , Cell Line , Chromatin/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , DNA-Binding Proteins/metabolism , E1A-Associated p300 Protein , Early Growth Response Protein 1 , Enzyme-Linked Immunosorbent Assay , Fixatives/pharmacology , Formaldehyde/pharmacology , Humans , Luciferases/metabolism , Macrophages/metabolism , Mice , Models, Biological , Molecular Sequence Data , Monocytes/microbiology , Plasmids/metabolism , Precipitin Tests , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets , Sp1 Transcription Factor/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/microbiology , Time Factors , Transcription Factors/metabolism , Transcription, Genetic , Transfection , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , ets-Domain Protein Elk-1
16.
Mol Cell Biol ; 22(8): 2620-31, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11909956

ABSTRACT

We present evidence that the inducer-specific regulation of the human tumor necrosis factor alpha (TNF-alpha) gene in T cells involves the assembly of distinct higher-order transcription enhancer complexes (enhanceosomes), which is dependent upon inducer-specific helical phasing relationships between transcription factor binding sites. While ATF-2, c-Jun, and the coactivator proteins CBP/p300 play a central role in TNF-alpha gene activation stimulated by virus infection or intracellular calcium flux, different sets of activators including NFATp, Sp1, and Ets/Elk are recruited to a shared set of transcription factor binding sites depending upon the particular stimulus. Thus, these studies demonstrate that the inducer-specific assembly of unique enhanceosomes is a general mechanism by which a single gene is controlled in response to different extracellular stimuli.


Subject(s)
Nuclear Proteins , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/genetics , Base Sequence , Binding Sites/genetics , Cell Line , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation , Humans , Models, Biological , Molecular Sequence Data , NFATC Transcription Factors , Promoter Regions, Genetic , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...