Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Cytokine ; 110: 110-115, 2018 10.
Article in English | MEDLINE | ID: mdl-29729649

ABSTRACT

Interstitial cystitis (IC) is a chronic syndrome that affects the urinary bladder. The etiology of this disease is unclear, and no effective therapies are available at this time. Although inflammation is suspected, no clear evidence for a role of conventional mediators of inflammation, such as cytokines and their downstream molecules, has been obtained to date. Our previous studies indicated that primary cell cultures derived from IC urothelium abnormally express molecules associated with cell adhesion. Here we describe a mechanism by which transcriptional changes in tight junction and adhesion molecules are mediated. Oncosuppressor proteins p53 and cyclin-dependent protein kinase inhibitor p21 directly associate with regulatory sites on the ZO-1 and E-cadherin genes, identifying important roles for p53 and p21 in driving non-oncogenic pathologies. These data also suggest that interference with these factors offers a potential therapeutic opportunity.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cystitis, Interstitial/metabolism , Gene Expression/physiology , Tumor Suppressor Protein p53/metabolism , Cadherins/metabolism , Cell Adhesion/physiology , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/physiology , Cell Line , Cytokines/metabolism , Humans , Inflammation/metabolism , Tight Junctions/metabolism , Tight Junctions/physiology , Transcription, Genetic/physiology , Urinary Bladder/metabolism , Urinary Bladder/physiology , Urothelium/metabolism , Zonula Occludens-1 Protein/metabolism
2.
Oncotarget ; 8(40): 68191-68207, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28978108

ABSTRACT

Studies suggest that tunicamycin may work as a therapeutic drug to cancer cells by inducing stress in the endoplasmic reticulum (ER) through unfolded protein response (UPR) and thereby promoting apoptosis. However, mechanisms of the prolonged activation of the UPR under sustained ER stress in the regulation of cell apoptosis are largely unknown. To delineate the role of candidate genes in the apoptotic process under ER stress and to search for new therapeutic strategies to treat metastatic castration resistant prostate cancer, we performed whole genome expression microarray analysis in tunicamycin treated metastatic androgen-insensitive prostate cancer cells, PC-3. Among several induced genes, the expression of eNOS (NOS3) gene was remarkably high. The increased expression of eNOS activates mTORC1 through RagC. This results into an accumulation of p62 (SQSTM1) which facilitates aggregation of ubiquitinated protein thus compromising clearance of misfolded toxic protein aggregates. Lastly, association of p62 proteins and misfolded proteins promote reactive oxygen species (ROS) mediated mitochondrial apoptosis. Overall, our data demonstrate that tunicamycin induced ER stress promotes prostate cancer cell death by activating mTORC1 through eNOS-RagC pathway.

3.
J Biol Chem ; 291(42): 22030-22042, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27590344

ABSTRACT

Expression of DAPK1, a critical regulator of autophagy and apoptosis, is lost in a wide variety of tumors, although the mechanisms are unclear. A transcription factor complex consisting of ATF6 (an endoplasmic reticulum-resident factor) and C/EBP-ß is required for the IFN-γ-induced expression of DAPK1 IFN-γ-induced proteolytic processing of ATF6 and phosphorylation of C/EBP-ß are obligatory for the formation of this transcriptional complex. We report that defects in this pathway fail to control growth of chronic lymphocytic leukemia (CLL). Consistent with these observations, IFN-γ and chemotherapeutics failed to activate autophagy in CLL patient samples lacking ATF6 and/or C/EBP-ß. Together, these results identify a molecular basis for the loss of DAPK1 expression in CLL.


Subject(s)
Activating Transcription Factor 6/metabolism , Autophagy , CCAAT-Enhancer-Binding Protein-beta/metabolism , Death-Associated Protein Kinases/biosynthesis , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Leukemic , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Neoplasm Proteins/metabolism , Activating Transcription Factor 6/genetics , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Line, Transformed , Death-Associated Protein Kinases/genetics , Female , Humans , Interferon-gamma/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Male , Neoplasm Proteins/genetics
4.
Pathog Dis ; 73(9): ftv080, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26424808

ABSTRACT

Bacillus anthracis, the causative agent of anthrax, is acquired by mammalian hosts from the environment, as quiescent endospores. These endospores must germinate inside host cells, forming vegetative bacilli, before they can express the virulence factors that enable them to evade host defenses and disseminate throughout the body. While the role of macrophages and dendritic cells in this initial interaction has been established, the role of polymorphonuclear leukocytes (PMNs) has not been adequately defined. We discovered that while B. anthracis 34F2 Sterne endospores germinate poorly within non-activated human PMNs, these phagocytes exhibit rapid microbicidal activity toward the outgrown vegetative bacilli, independent of superoxide and nitric oxide. These findings suggest that a non-free radical pathway kills B. anthracis bacilli. We also find in PMNs an autophagic mechanism of bacterial killing based on the rapid induction of LC-3 conversion, beclin-1 expression, sequestosome 1 (SQSTM1) degradation and inhibition of bactericidal activity by the inhibitor, 3-methyladenine. These findings extend to PMNs an autophagic bactericidal mechanism previously described for other phagocytes.


Subject(s)
Anthrax/immunology , Anthrax/pathology , Autophagy , Bacillus anthracis/immunology , Microbial Viability , Neutrophils/immunology , Neutrophils/microbiology , Animals , Blotting, Western , Cells, Cultured , Humans , Mice , Phagocytosis , Reactive Nitrogen Species/analysis , Reactive Oxygen Species/analysis
5.
Mol Cell Biol ; 34(21): 4033-48, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25135476

ABSTRACT

The death-associated protein kinase 1 (DAPK1) is an important regulator of cell death and autophagy. Recently, we have identified that ATF6, an endoplasmic reticulum-resident transcription factor, in association with the transcription factor CEBP-ß, regulates the gamma interferon (IFN-γ)-induced expression of Dapk1 (P. Gade et al., Proc. Natl. Acad. Sci. U. S. A. 109:10316-10321, 2012, doi.org/10.1073/pnas.1119273109). IFN-γ-induced proteolytic processing of ATF6 and phosphorylation of C/EBP-ß were essential for the formation of a novel transcriptional complex that regulates DAPK1. Here, we report that IFN-γ activates the ASK1-MKK3/MKK6-p38 mitogen-activated protein kinase (MAPK) pathway for controlling the activity of ATF6. The terminal enzyme in this pathway, p38 MAPK, phosphorylates a critical threonine residue in ATF6 upstream of its DNA binding domain. ATF6 mutants defective for p38 MAPK phosphorylation fail to undergo proteolytic processing in the Golgi apparatus and drive IFN-γ-induced gene expression and autophagy. We also show that mice lacking Ask1 are highly susceptible to lethal bacterial infection owing to defective autophagy. Together, these results identify a novel host defense pathway controlled by IFN-γ signaling.


Subject(s)
Activating Transcription Factor 6/metabolism , Autophagy , Death-Associated Protein Kinases/metabolism , Interferon-gamma/metabolism , MAP Kinase Kinase Kinase 5/metabolism , MAP Kinase Signaling System , Activating Transcription Factor 6/genetics , Animals , Bacillus anthracis/pathogenicity , Binding Sites , Cell Line , Death-Associated Protein Kinases/genetics , Gene Knockdown Techniques , Humans , Liver/immunology , Liver/microbiology , Mice , Mutation , Phosphorylation , Sf9 Cells , Spleen/immunology , Spleen/microbiology
6.
Am J Infect Control ; 42(3): e33-6, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24581026

ABSTRACT

BACKGROUND: Streamlining health care-associated infection surveillance is essential for health care facilities owing to the continuing increases in reporting requirements. METHODS: Stanford Hospital, a 583-bed adult tertiary care center, used their electronic medical record (EMR) to develop an electronic algorithm to reduce the time required to conduct catheter-associated urinary tract infection (CAUTI) surveillance in adults. The algorithm provides inclusion and exclusion criteria, using the National Healthcare Safety Network definitions, for patients with a CAUTI. The algorithm was validated by trained infection preventionists through complete chart review for a random sample of cultures collected during the study period, September 1, 2012, to February 28, 2013. RESULTS: During the study period, a total of 6,379 positive urine cultures were identified. The Stanford Hospital electronic CAUTI algorithm identified 6,101 of these positive cultures (95.64%) as not a CAUTI, 191 (2.99%) as a possible CAUTI requiring further validation, and 87 (1.36%) as a definite CAUTI. Overall, use of the algorithm reduced CAUTI surveillance requirements at Stanford Hospital by 97.01%. CONCLUSIONS: The electronic algorithm proved effective in increasing the efficiency of CAUTI surveillance. The data suggest that CAUTI surveillance using the National Healthcare Safety Network definitions can be fully automated.


Subject(s)
Catheter-Related Infections/epidemiology , Electronic Health Records , Epidemiological Monitoring , Urinary Tract Infections/epidemiology , California/epidemiology , Electronic Data Processing , Hospitals , Humans
7.
Breast Cancer Res Treat ; 145(1): 5-22, 2014 May.
Article in English | MEDLINE | ID: mdl-24668500

ABSTRACT

Nicotine, a main addictive compound in tobacco smoke, has been linked to promotion and progression of lung, head and neck, pancreatic, and breast cancers, but the detailed mechanisms of cancer progression remain elusive. Here, we show that nicotine induces the expression of galectin-3 (an anti-apoptotic ß-galactoside-binding lectin) in breast cancer cell line and in primary tumors from breast cancer patients. Nicotine-induced up regulation of galectin-3 is due to an increased expression of α9 isoform of nicotinic acetylcholine receptor (α9nAChR), which activates transcription factor STAT3 that in turn, physically binds to galectin-3 (LGALS3) promoter and induces transcription of galectin-3. Intracellular galectin-3 increased mitochondrial integrity and suppressed chemotherapeutic-induced apoptosis of breast cancer cell. Moreover, nicotine-induced enrichment of side population cells with cancer stem cell-like properties was modulated by galectin-3 expression and could be significantly reduced by transient knock down of LGALS3 and its upstream signaling molecules STAT3 and α9nAChR. Thus, galectin-3 or its upstream signaling molecule STAT3 or α9nAChR could be a potential target to prevent nicotine-induced chemoresistance in breast cancer.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Signal Transduction/drug effects , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Fragmentation , Galectin 3/biosynthesis , Humans , Immunohistochemistry , Neoplastic Stem Cells/pathology , Real-Time Polymerase Chain Reaction , Receptors, Nicotinic/metabolism , STAT3 Transcription Factor/metabolism , Side-Population Cells , Transfection
8.
PLoS One ; 8(3): e57855, 2013.
Article in English | MEDLINE | ID: mdl-23472114

ABSTRACT

SerpinB2 or plasminogen activator inhibitor type 2 (PAI-2) is highly induced in macrophages in response to inflammatory stimuli and is linked to the modulation of innate immunity, macrophage survival, and inhibition of plasminogen activators. Lipopolysaccharide (LPS), a potent bacterial endotoxin, can induce SerpinB2 expression via the toll-like receptor 4 (TLR4) by ∼1000-fold over a period of 24 hrs in murine macrophages. To map the LPS-regulated SerpinB2 promoter regions, we transfected reporter constructs driven by the ∼5 kb 5'-flanking region of the murine SerpinB2 gene and several deletion mutants into murine macrophages. In addition, we compared the DNA sequence of the murine 5' flanking sequence with the sequence of the human gene for homologous functional regulatory elements and identified several regulatory cis-acting elements in the human SERPINB2 promoter conserved in the mouse. Mutation analyses revealed that a CCAAT enhancer binding (C/EBP) element, a cyclic AMP response element (CRE) and two activator protein 1 (AP-1) response elements in the murine SerpinB2 proximal promoter are essential for optimal LPS-inducibility. Electrophoretic mobility shift (EMSA) and chromatin immunoprecipitation (ChIP) assays demonstrated that LPS induces the formation of C/EBP-ß containing complexes with the SerpinB2 promoter. Importantly, both constitutive and LPS-induced SerpinB2 expression was severely abrogated in C/EBP-ß-null mouse embryonic fibroblasts (MEFs) and primary C/EBP-ß-deficient peritoneal macrophages. Together, these data provide new insight into C/EBP-ß-dependent regulation of inflammation-associated SerpinB2 expression.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Gene Expression Regulation , Lipopolysaccharides/metabolism , Plasminogen Activator Inhibitor 2/metabolism , Animals , Base Sequence , Cell Line , Conserved Sequence , DNA Mutational Analysis , Fibroblasts/cytology , Humans , Macrophages/metabolism , Mice , Molecular Sequence Data , Promoter Regions, Genetic
9.
Autophagy ; 8(11): 1673-4, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22874566

ABSTRACT

IFNG/IFNγ plays a critical role in driving innate and acquired defenses against infectious pathogens. The death-associated protein kinase 1 (DAPK1), originally identified as an activator of IFNG-induced cell death, controls autophagy. Previously, we have shown that transcription factor CEBPB (C/EBP-ß) regulates IFNG-induced expression of Dapk1 through a CRE/ATF motif in its enhancer. In this paper we have shown that ATF6, an ER-resident transcription factor regulates IFNG-induced Dapk1 expression through the CRE/ATF site, in association with CEBPB. IFNG-stimulated proteolytic cleavage of ATF6, and MAPK1/3 (ERK2/1)-dependent phosphorylation of CEBPB together control the expression of Dapk1. Consistent with their requirement for DAPK1 expression, IFNG fails to induce autophagy in cells lacking either Atf6 or Cebpb. More importantly, the Atf6(-/-) mice are highly susceptible to lethal bacterial infections due to a loss of autophagy. This study reported a connection between ER stress and autophagy in mediating antibacterial defenses.


Subject(s)
Activating Transcription Factor 6/metabolism , Autophagy , Bacterial Infections/pathology , Bacterial Infections/prevention & control , Endoplasmic Reticulum Stress , Interferon-gamma/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Bacterial Infections/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Death-Associated Protein Kinases , Humans , Mice , Models, Biological
10.
Proc Natl Acad Sci U S A ; 109(26): 10316-21, 2012 Jun 26.
Article in English | MEDLINE | ID: mdl-22699507

ABSTRACT

The IFN family of cytokines operates a frontline defense against pathogens and neoplastic cells in vivo by controlling the expression of several genes. The death-associated protein kinase 1 (DAPK1), an IFN-γ-induced enzyme, controls cell cycle, apoptosis, autophagy, and tumor metastasis, and its expression is frequently down-regulated in a number of human tumors. Although the biochemical action of DAPK1 is well understood, mechanisms that regulate its expression are unclear. Previously, we have shown that transcription factor C/EBP-ß is required for the basal and IFN-γ-induced expression of DAPK1. Here, we show that ATF6, an ER stress-induced transcription factor, interacts with C/EBP-ß in an IFN-stimulated manner and is obligatory for Dapk1 expression. IFN-stimulated proteolytic processing of ATF6 and ERK1/2-mediated phosphorylation of C/EBP-ß are necessary for these interactions. More importantly, IFN-γ failed to activate autophagic response in cells lacking either ATF6 or C/EBP-ß. Consistent with these observations, the Atf6(-/-) mice were highly susceptible to lethal bacterial infections compared with the wild-type mice. These studies not only unravel an IFN signaling pathway that controls cell growth and antibacterial defense, but also expand the role of ATF6 beyond ER stress.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Autophagy/physiology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Interferon-gamma/physiology , Animals , Apoptosis Regulatory Proteins/genetics , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Death-Associated Protein Kinases , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Proteolysis , Transcription Factors/metabolism
11.
Clin Cancer Res ; 18(2): 360-369, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22096027

ABSTRACT

PURPOSE: Death-associated protein kinase 1 (DAPK1), a tumor suppressor, is a rate-limiting effector in an endoplasmic reticulum (ER) stress-dependent apoptotic pathway. Its expression is epigenetically suppressed in several tumors. A mechanistic basis for epigenetic/transcriptional repression of DAPK1 was investigated in certain forms of acute myeloid leukemia (AML) with poor prognosis, which lacked ER stress-induced apoptosis. EXPERIMENTAL DESIGN: Heterogeneous primary AMLs were screened to identify a subgroup with Flt3ITD in which repression of DAPK1, among NF-κB-and c-Jun-responsive genes, was studied. RNA interference knockdown studies were carried out in an Flt3ITD(+) cell line, MV-4-11, to establish genetic epistasis in the pathway Flt3ITD-TAK1-DAPK1 repression, and chromatin immunoprecipitations were carried out to identify proximate effector proteins, including TAK1-activated p52NF-κB, at the DAPK1 locus. RESULTS: AMLs characterized by normal karyotype with Flt3ITD were found to have 10- to 100-fold lower DAPK1 transcripts normalized to the expression of c-Jun, a transcriptional activator of DAPK1, as compared with a heterogeneous cytogenetic category. In addition, Meis1, a c-Jun-responsive adverse AML prognostic gene signature was measured as control. These Flt3ITD(+) AMLs overexpress relB, a transcriptional repressor, which forms active heterodimers with p52NF-κB. Chromatin immunoprecipitation assays identified p52NF-κB binding to the DAPK1 promoter together with histone deacetylase 2 (HDAC2) and HDAC6 in the Flt3ITD(+) human AML cell line MV-4-11. Knockdown of p52NF-κB or its upstream regulator, NF-κB-inducing kinase (NIK), de-repressed DAPK1. DAPK1-repressed primary Flt3ITD(+) AMLs had selective nuclear activation of p52NF-κB. CONCLUSIONS: Flt3ITD promotes a noncanonical pathway via TAK1 and p52NF-κB to suppress DAPK1 in association with HDACs, which explains DAPK1 repression in Flt3ITD(+) AML.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Leukemia, Myeloid, Acute/metabolism , NF-kappa B p52 Subunit/metabolism , Signal Transduction , fms-Like Tyrosine Kinase 3/metabolism , Apoptosis , Apoptosis Regulatory Proteins/genetics , Binding Sites , Calcium-Calmodulin-Dependent Protein Kinases/genetics , DNA Methylation , Death-Associated Protein Kinases , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation, Leukemic , HL-60 Cells , Humans , Karyotype , Leukemia, Myeloid, Acute/genetics , MAP Kinase Kinase Kinases/metabolism , Mutation , Promoter Regions, Genetic , Protein Binding , Tandem Repeat Sequences , Tumor Cells, Cultured , fms-Like Tyrosine Kinase 3/genetics
12.
Methods Mol Biol ; 809: 85-104, 2012.
Article in English | MEDLINE | ID: mdl-22113270

ABSTRACT

Differential gene expression is facilitated by transcriptional regulatory mechanisms and chromatin modifications through DNA-protein interactions. One of the widely used assays to study this is chromatin immunoprecipitation (ChIP) assay, which enables analysis of association of regulatory molecules to specific promoters and histone modifications in vivo. This is of immense value as ChIP assays can provide glimpse of the regulatory mechanisms involved in gene expression in vivo. This article outlines the general strategies and protocols to study ChIP assays in differential recruitment of transcriptional factors (TFs) and also global analysis of transcription factor recruitment is discussed. Further, the applications of ChIP assays for discovering novel genes that are dependent on specific transcription factors were addressed.


Subject(s)
Chromatin Immunoprecipitation/methods , Transcription Factors/metabolism , Animals , Blotting, Western , Cells, Cultured , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Immunoprecipitation , Mice , Protein Binding , Transcription Factors/genetics
13.
PLoS One ; 6(9): e24082, 2011.
Article in English | MEDLINE | ID: mdl-21931644

ABSTRACT

We have recently isolated novel IFN-inducible gene, Gene associated with Retinoid-Interferon-induced Mortality-1 (GRIM-1), using a genetic technique. Moderate ectopic expression of GRIM-1 caused growth inhibition and sensitized cells to retinoic acid (RA)/IFN-induced cell death while high expression caused apoptosis. GRIM-1 depletion, using RNAi, conferred a growth advantage. Three protein isoforms (1α, 1ß and 1γ) with identical C-termini are produced from GRIM-1 mRNA. We show that GRIM-1 isoforms interact with NAF1 and DKC1, two essential proteins required for box H/ACA sno/sca RNP biogenesis and suppresses box H/ACA RNA levels in mammalian cells by delocalizing NAF1. Suppression of these small RNAs manifests as inefficient rRNA maturation and growth suppression. Interestingly, yeast Shq1p also caused growth suppression in mammalian cells. Consistent with its growth-suppressive property, GRIM-1 expression is lost in a number of human primary prostate tumors. Our observations support a recent study that GRIM-1 might act as a co-tumor suppressor in the prostate.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Gene Expression Regulation, Neoplastic , RNA, Ribosomal/genetics , RNA, Small Nucleolar/genetics , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Growth Processes/drug effects , Cell Growth Processes/genetics , Cell Line, Tumor , HeLa Cells , Humans , Immunohistochemistry , Interferon-beta/pharmacology , Male , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Ribosomal/metabolism , RNA, Small Nucleolar/metabolism , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Tretinoin/pharmacology
14.
J Immunol ; 183(3): 1657-66, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19592659

ABSTRACT

CD1d is a nonclassical Ag-presenting molecule that presents glycolipid Ags to NKT cells that are involved in immune defense and tumor rejection. It also plays a role in immunoregulatory functions in the epidermis. The mechanisms controlling the expression of CD1d are not well understood. Therefore, we cloned the CD1d gene promoter and characterized its activities in primary human keratinocytes and other cell lines of epithelial origin. We found that a CCAAT box in the CD1d promoter is required for its expression in keratinocytes. We show here that transcription factor C/EBP-beta binds to the CCAAT box in the CD1d promoter in vitro and in vivo. Consistent with these observations, deletion of the gene encoding for C/EBP-beta caused a loss of CD1d expression. The in vivo regulation of CD1d has significant implications for the pathologic mechanisms of certain immunologic skin diseases in which NKT cells play a role, such as allergic contact dermatitis and psoriasis. Together, these data show a central role for C/EBP-beta in regulating CD1d transcription.


Subject(s)
Antigens, CD1d/genetics , CCAAT-Enhancer-Binding Protein-beta/physiology , Gene Expression Regulation , Keratinocytes/metabolism , Transcription, Genetic , Binding Sites , Cell Line , Cloning, Molecular , Epithelial Cells , Humans , Natural Killer T-Cells , Promoter Regions, Genetic , Skin Diseases/etiology , Skin Diseases/immunology
15.
Int J Cancer ; 125(7): 1566-74, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19521987

ABSTRACT

DAPK1, a ca(+2)/calmodulin regulated serine/threonine kinase, is a major tumor suppressor, whose expression is lost in multiple tumor types. However, the mechanisms contributing to it are unclear. We have recently shown that CCAAT/Enhancer binding protein-beta (C/EBP-beta) is required for the basal and interferon gamma (IFN-gamma)-induced expression of dapk1 in many cell types. C/EBP-beta interacts with the transcriptional Mediator, a multisubunit complex that couples enhancer bound transcription factors to the basal transcriptional machinery in an IFN-gamma dependent manner for regulating dapk1 expression. Specifically, the Med1 (TRAP220/PBP/DRIP220/CRSP220) subunit associates with the enhancer bound C/EBP-beta at the CRE/ATF site of dapk1 in an IFN-gamma dependent manner for stimulating gene expression. Therefore, we investigated if the mechanism responsible for the loss of dapk1 expression in human cancers involves a failure to recruit C/EBP-beta and/or Med1 to the dapk1 promoter. We compared the relative occupancy of these factors at the dapk1 promoter at CRE/ATF sites in normal and cancer cell lines. A significantly lower binding of these factors to the CRE/ATF site of dapk1 promoter occurred in human cancer cell lines than in normal cells. We show that loss of Med1 expression correlates with a corresponding loss of dapk1 expression in a number of primary human lung carcinomas. Med1 levels were significantly lower in cancer cell lines than in normal controls. Importantly, we show that restoration of Med1 induces the expression of dapk1 in these cancer cells and also attenuates their metastatic potential in vivo. Our studies reveal a critical parameter limiting dapk1 expression in cancer cell lines.


Subject(s)
Adenocarcinoma/metabolism , Apoptosis Regulatory Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Interferon-gamma/metabolism , Lung Neoplasms/metabolism , Transcription Factors/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Death-Associated Protein Kinases , Down-Regulation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Mediator Complex Subunit 1 , Mice , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous
16.
Sci Signal ; 2(59): ra8, 2009 Feb 24.
Article in English | MEDLINE | ID: mdl-19244213

ABSTRACT

Interleukin-17 (IL-17), the hallmark cytokine of T helper 17 (T(H)17) cells, signals through a distinct receptor subclass, yet little is known about the mechanisms involved. IL-17 activates the expression of target genes through the actions of the transcription factors nuclear factor kappaB (NF-kappaB), CAAT enhancer binding protein delta (C/EBPdelta), and C/EBPbeta. The adaptor proteins tumor necrosis factor receptor-associated factor 6 (TRAF6) and Act1 are upstream of NF-kappaB and C/EBPdelta, but the regulation of C/EBPbeta remains undefined. Here, we show that IL-17 signaling led to phosphorylation of two sites in the regulatory 2 domain of C/EBPbeta in a sequential, interdependent fashion. The first was rapid and dependent on extracellular signal-regulated kinase (ERK), whereas the second was dependent on the activity of glycogen synthase kinase 3beta (GSK-3beta). These pathways were mediated by distinct subdomains within IL-17 receptor A (IL-17RA). Whereas phosphorylation of threonine 188 (Thr188) was mediated by the previously identified SEF/IL-17R homology domain-Toll-IL-1R-like loop (SEFIR-TILL), phosphorylation of Thr179 occurred through a newly characterized motif located in the distal tail of IL-17RA. Phosphorylated C/EBPbeta mediated a negative signal, because blocking ERK and GSK-3beta increased expression of IL-17 target genes and a C/EBPbeta-Thr188 mutant enhanced activation of a C/EBP-dependent reporter. Overexpression of GSK-3beta inhibited IL-17-induced activation of a C/EBP-dependent reporter, and Thr179 of C/EBPbeta was not phosphorylated in GSK-3beta-deficient cells. Thus, IL-17 triggered the dual phosphorylation of C/EBPbeta, which inhibited the expression of proinflammatory genes. This detailed dissection is the first for the IL-17-mediated C/EBP pathway and the first known example of a negative signal mediated by IL-17RA.


Subject(s)
CCAAT-Enhancer-Binding Protein-delta/metabolism , Models, Biological , Protein Structure, Tertiary/physiology , Receptors, Interleukin-17/metabolism , Signal Transduction/physiology , Blotting, Western , Immunoprecipitation , Luciferases , NF-kappa B/metabolism , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction , Tandem Mass Spectrometry
17.
J Biol Chem ; 283(19): 13077-86, 2008 May 09.
Article in English | MEDLINE | ID: mdl-18339625

ABSTRACT

Transcription factor CCAAT/enhancer-binding protein (C/EBP)-beta is crucial for regulating transcription of genes involved in a number of diverse cellular processes, including those involved in some cytokine-induced responses. However, the mechanisms that contribute to its diverse transcriptional activity are not yet fully understood. To gain an understanding into its mechanisms of action, we took a proteomic approach and identified cellular proteins that associate with C/EBP-beta in an interferon (IFN)-gamma-dependent manner. Transcriptional mediator (Mediator) is a multisubunit protein complex that regulates signal-induced cellular gene transcription from enhancer-bound transcription factor(s). Here, we report that the Med1 subunit of the Mediator as a C/EBP-beta-interacting protein. Using gene knock-out cells and mutational and RNA interference approaches, we show that Med1 is critical for IFN-induced expression of certain genes. Med1 associates with C/EBP-beta through a domain located between amino acids 125 and 155 of its N terminus. We also show that the MAPK, ERK1/2, and an ERK phosphorylation site within regulatory domain 2, more specifically the Thr(189) residue, of C/EBP-beta are essential for it to bind to Med1. Last, an ERK-regulated site in Med1 protein is also essential for up-regulating IFN-induced transcription although not critical for binding to C/EBP-beta.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Endodeoxyribonucleases/metabolism , Interferon-gamma/pharmacology , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , CCAAT-Enhancer-Binding Protein-beta/deficiency , CCAAT-Enhancer-Binding Protein-beta/genetics , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , Death-Associated Protein Kinases , Endodeoxyribonucleases/chemistry , Endodeoxyribonucleases/genetics , Gene Expression Regulation/drug effects , Humans , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Protein Structure, Tertiary , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Proteins , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
18.
Mol Cell Biol ; 28(8): 2528-48, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18250155

ABSTRACT

Transcription factor C/EBP-beta regulates a number of physiological responses. During an investigation of the growth-suppressive effects of interferons (IFNs), we noticed that cebpb(-/-) cells fail to undergo apoptosis upon gamma IFN (IFN-gamma) treatment, compared to wild-type controls. To examine the basis for this response, we have performed gene expression profiling of isogenic wild-type and cebpb(-/-) bone marrow macrophages and identified a number of IFN-gamma-regulated genes that are dependent on C/EBP-beta for their expression. These genes are distinct from those regulated by the JAK-STAT pathways. Genes identified in this screen appear to participate in various cellular pathways. Thus, we identify a new pathway through which the IFNs exert their effects on cellular genes through C/EBP-beta. One of these genes is death-associated protein kinase 1 (dapk1). DAPK1 is critical for regulating the cell cycle, apoptosis, and metastasis. Using site-directed mutagenesis, RNA interference, and chromatin immunoprecipitation assays, we show that C/EBP-beta binds to the promoter of dapk1 and is required for the regulation of dapk1. Both mouse dapk1 and human dapk1 exhibited similar dependences on C/EBP-beta for their expression. The expression of the other members of the DAPK family occurred independently of C/EBP-beta. Members of the C/EBP family of transcription factors other than C/EBP-beta did not significantly affect dapk1 expression. We identified two elements in this promoter that respond to C/EBP-beta. One of these is a consensus C/EBP-beta-binding site that constitutively binds to C/EBP-beta. The other element exhibits homology to the cyclic AMP response element/activating transcription factor binding sites. C/EBP-beta binds to this site in an IFN-gamma-dependent manner. Inhibition of ERK1/2 or mutation of an ERK1/2 site in the C/EBP-beta protein suppressed the IFN-gamma-induced response of this promoter. Together, our data show a critical role for C/EBP-beta in a novel IFN-induced cell growth-suppressive pathway via DAPK1.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Gene Expression Regulation , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/classification , Apoptosis Regulatory Proteins/genetics , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , CCAAT-Enhancer-Binding Protein-beta/deficiency , CCAAT-Enhancer-Binding Protein-beta/genetics , Calcium-Calmodulin-Dependent Protein Kinases/classification , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cell Line , Death-Associated Protein Kinases , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Humans , Interferon-gamma/pharmacology , MAP Kinase Signaling System/drug effects , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Knockout , Promoter Regions, Genetic
19.
Cell Mol Immunol ; 4(6): 407-18, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18163952

ABSTRACT

Cytokines like interferons (IFNs) play a central role in regulating innate and specific immunities against the pathogens and neoplastic cells. A number of signaling pathways are induced in response to IFN in various cells. One classic mechanism employed by IFNs is the JAK-STAT signaling pathway for inducing cellular responses. Here we describe the non-STAT pathways that participate in IFN-induced responses. In particular, we will focus on the role played by transcription factor C/EBP-beta in mediating these responses.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta , Interferons , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Signal Transduction/immunology , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/immunology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Humans , Immunity, Innate , Immunotherapy/trends , Intercellular Signaling Peptides and Proteins/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Interferons/immunology , Interferons/metabolism , Janus Kinases/immunology , MAP Kinase Signaling System/immunology , Paracrine Communication/immunology , STAT Transcription Factors/immunology , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...