Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 10(9): 949-57, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19668222

ABSTRACT

All metazoan guts are in permanent contact with the microbial realm. However, understanding of the exact mechanisms by which the strength of gut immune responses is regulated to achieve gut-microbe mutualism is far from complete. Here we identify a signaling network composed of complex positive and negative mechanisms that controlled the expression and activity of dual oxidase (DUOX), which 'fine tuned' the production of microbicidal reactive oxygen species depending on whether the gut encountered infectious or commensal microbes. Genetic analyses demonstrated that negative and positive regulation of DUOX was required for normal host survival in response to colonization with commensal and infectious microbes, respectively. Thus, the coordinated regulation of DUOX enables the host to achieve gut-microbe homeostasis by efficiently combating infection while tolerating commensal microbes.


Subject(s)
Drosophila/immunology , NADPH Oxidases/physiology , Activating Transcription Factor 2/physiology , Animals , Caco-2 Cells , Calcineurin/physiology , Carrier Proteins/physiology , Gene Expression Regulation, Enzymologic , Humans , Intestines/immunology , Intestines/microbiology , MAP Kinase Kinase 3/physiology , MAP Kinase Kinase Kinase 1/physiology , NADPH Oxidases/genetics , Phospholipase C beta/physiology , Reactive Oxygen Species/metabolism , Signal Transduction , Transcription, Genetic , p38 Mitogen-Activated Protein Kinases/physiology
2.
Lab Invest ; 98(5): 670-681, 2018 05.
Article in English | MEDLINE | ID: mdl-29382922

ABSTRACT

Rho guanine nucleotide exchange factor 39 (ARHGEF39), also called C9orf100, is a new member of the Dbl-family of guanine nucleotide exchange factors. Although ARHGEF39 has been proven to regulate tumor progression in hepatocellular carcinoma, the downstream signaling pathway of ARHGEF39 and its clinical associations in non-small cell lung cancer (NSCLC) are currently unknown. In the present study, using MTT, colony formation, flow cytometry, mice xenografts, wound healing, and transwell assays, we showed that ARHGEF39 promoted tumor proliferation, migration, and invasion. Furthermore, ARHGEF39 promoted the expression of Cyclin A2, Cyclin D1, and MMP2 by activating Rac1, leading to increased phosphorylation of P38 and ATF2. Treatment with a P38 inhibitor counteracted the effect of ARHGEF39 overexpression on the increase in Cyclin A2, Cyclin D1, and MMP2 expression. Moreover, the elevated levels of p-P38 and p-ATF2 caused by ARHGEF39 overexpression could be inhibited by expression of a dominant negative Rac1 mutant (T17N). In addition, the inhibition of the expression of p-P38 and p-ATF2 by ARHGEF39 RNAi could be restored by the expression of a constitutively active Rac1 mutant (Q61L). A similar impact on cell growth and invasion was observed after ARHGEF39 overexpression combined with the P38 inhibitor, Rac1 T17N, or Rac1 Q61L. Using immunohistochemistry, ARHGEF39 expression was observed to correlate positively with larger tumor size in clinical samples from 109 cases of NSCLC (P = 0.008). The Kaplan-Meier test revealed that ARHGEF39 expression significantly affected the overall survival of patients with NSCLC (52.55 ± 6.40 months vs. 64.30 ± 5.40 months, P = 0.017). In conclusion, we identified that ARHGEF39 promotes tumor growth and invasion by activating the Rac1-P38-ATF2 signaling pathway, as well as increasing the expression of Cyclin A2, Cyclin D1, and MMP2 in NSCLC cells. ARHGEF39 may be a useful marker to predict poor prognosis of patients with NSCLC.


Subject(s)
Activating Transcription Factor 2/physiology , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Rho Guanine Nucleotide Exchange Factors/physiology , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/physiology , rac1 GTP-Binding Protein/physiology , Adult , Aged , Animals , Carcinoma, Non-Small-Cell Lung/mortality , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation , Female , Humans , Lung Neoplasms/mortality , Mice , Mice, Inbred BALB C , Middle Aged , Phosphorylation , Prognosis
3.
J Immunol ; 189(2): 1014-23, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22706082

ABSTRACT

Low-dose endotoxemia is prevalent in humans with adverse health conditions, and it correlates with the pathogenesis of chronic inflammatory diseases such as atherosclerosis, diabetes, and neurologic inflammation. However, the underlying molecular mechanisms are poorly understood. In this study, we demonstrate that subclinical low-dose LPS skews macrophages into a mild proinflammatory state, through cell surface TLR4, IL-1R-associated kinase-1, and the Toll-interacting protein. Unlike high-dose LPS, low-dose LPS does not induce robust activation of NF-κB, MAPKs, PI3K, or anti-inflammatory mediators. Instead, low-dose LPS induces activating transcription factor 2 through Toll-interacting protein-mediated generation of mitochondrial reactive oxygen species, allowing mild induction of proinflammatory mediators. Low-dose LPS also suppresses PI3K and related negative regulators of inflammatory genes. Our data reveal novel mechanisms responsible for skewed and persistent low-grade inflammation, a cardinal feature of chronic inflammatory diseases.


Subject(s)
Gene Expression Regulation/immunology , Inflammation Mediators/metabolism , Lipopolysaccharides/toxicity , Macrophages/immunology , Macrophages/pathology , Activating Transcription Factor 2/physiology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Cells, Cultured , Dose-Response Relationship, Immunologic , Inflammation Mediators/physiology , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/physiology , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondria/immunology , Mitochondria/pathology , Phosphatidylinositol 3-Kinase/physiology , Phosphoinositide-3 Kinase Inhibitors , Reactive Oxygen Species/metabolism , Signal Transduction/immunology
4.
Cancer Metastasis Rev ; 31(3-4): 621-32, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22684365

ABSTRACT

The progression of melanoma toward the metastatic phenotype occurs in a defined stepwise manner. While many molecular changes take place early in melanoma development, progression toward the malignant phenotype, most notably during the transition from the radial growth phase (RGP) to the vertical growth phase (VGP) involves deregulated expression of several transcription factors. For example, the switch from RGP to VGP is associated with the loss of the transcription factor AP2α and gain of transcriptional activity of cAMP-responsive element binding protein. Together with the upregulation of microphthalmia-associated transcription factor, activating transcription factor 2, nuclear factor kappa B, and other transcription factors, these changes lead to dysregulated expression or function of important cellular adhesion molecules, matrix degrading enzymes, survival factors, as well as other factors leading to metastatic melanoma. Additionally, recent evidence suggests that microRNAs and RNA editing machinery influence the expression of transcription factors or are regulated themselves by transcription factors. Many of the downstream signaling molecules regulated by transcription factors, such as protease activated receptor-1, interleukin-8, and MCAM/MUC18 represent new treatment prospects.


Subject(s)
Melanoma/secondary , Transcription Factors/physiology , Activating Transcription Factor 1/physiology , Activating Transcription Factor 2/physiology , Animals , Cyclic AMP Response Element-Binding Protein/physiology , Gene Expression Regulation, Neoplastic , Humans , Melanoma/drug therapy , Microphthalmia-Associated Transcription Factor/physiology , NF-kappa B/physiology , RNA Editing , Transcription Factor AP-2/physiology
5.
J Neuroinflammation ; 9: 110, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22643046

ABSTRACT

BACKGROUND: Lipoteichoic acid (LTA) is a component of gram-positive bacterial cell walls and may be elevated in the cerebrospinal fluid of patients suffering from meningitis. Among matrix metalloproteinases (MMPs), MMP-9 has been observed in patients with brain inflammatory diseases and may contribute to the pathology of brain diseases. Moreover, several studies have suggested that increased oxidative stress is implicated in the pathogenesis of brain inflammation and injury. However, the molecular mechanisms underlying LTA-induced redox signal and MMP-9 expression in brain astrocytes remain unclear. OBJECTIVE: Herein we explored whether LTA-induced MMP-9 expression was mediated through redox signals in rat brain astrocytes (RBA-1 cells). METHODS: Upregulation of MMP-9 by LTA was evaluated by zymographic and RT-PCR analyses. Next, the MMP-9 regulatory pathways were investigated by pretreatment with pharmacological inhibitors or transfection with small interfering RNAs (siRNAs), Western blotting, and chromatin immunoprecipitation (ChIP)-PCR and promoter activity reporter assays. Moreover, we determined the cell functional changes by migration assay. RESULTS: These results showed that LTA induced MMP-9 expression via a PKC(α)-dependent pathway. We further demonstrated that PKCα stimulated p47phox/NADPH oxidase 2 (Nox2)-dependent reactive oxygen species (ROS) generation and then activated the ATF2/AP-1 signals. The activated-ATF2 bound to the AP-1-binding site of MMP-9 promoter, and thereby turned on MMP-9 gene transcription. Additionally, the co-activator p300 also contributed to these responses. Functionally, LTA-induced MMP-9 expression enhanced astrocytic migration. CONCLUSION: These results demonstrated that in RBA-1 cells, activation of ATF2/AP-1 by the PKC(α)-mediated Nox(2)/ROS signals is essential for upregulation of MMP-9 and cell migration enhanced by LTA.


Subject(s)
Astrocytes/enzymology , Brain/enzymology , Lipopolysaccharides/physiology , Matrix Metalloproteinase 9/biosynthesis , NADPH Oxidases/physiology , Up-Regulation/physiology , Activating Transcription Factor 2/physiology , Animals , Animals, Newborn , Astrocytes/drug effects , Brain/cytology , Brain/drug effects , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Membrane Glycoproteins/physiology , NADPH Oxidase 2 , Oxidation-Reduction/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Teichoic Acids , Up-Regulation/drug effects
6.
Proc Natl Acad Sci U S A ; 105(5): 1674-9, 2008 Feb 05.
Article in English | MEDLINE | ID: mdl-18227516

ABSTRACT

Activating transcription factor 2 (ATF2) regulates transcription in response to stress and growth factor stimuli. Here, we use a mouse model in which ATF2 was selectively deleted in keratinocytes. Crossing the conditionally expressed ATF2 mutant with K14-Cre mice (K14.ATF2(f/f)) resulted in selective expression of mutant ATF2 within the basal layer of the epidermis. When subjected to a two-stage skin carcinogenesis protocol [7,12-dimethylbenz[a]anthracene/phorbol 12-tetradecanoate 13-acetate (DMBA/TPA)], K14.ATF2(f/f) mice showed significant increases in both the incidence and prevalence of papilloma development compared with the WT ATF2 mice. Consistent with these findings, keratinocytes of K14.ATF2(f/f) mice exhibit greater anchorage-independent growth compared with ATF2 WT keratinocytes. Papillomas of K14.ATF2(f/f) mice exhibit reduced expression of presenilin1, which is associated with enhanced beta-catenin and cyclin D1, and reduced Notch1 expression. Significantly, a reduction of nuclear ATF2 and increased beta-catenin expression were seen in samples of squamous and basal cell carcinoma, as opposed to normal skin. Our data reveal that loss of ATF2 transcriptional activity serves to promote skin tumor formation, thereby indicating a suppressor activity of ATF2 in skin tumor formation.


Subject(s)
Activating Transcription Factor 2/physiology , Papilloma/genetics , Skin Neoplasms/genetics , Tumor Suppressor Proteins/physiology , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Activating Transcription Factor 2/analysis , Activating Transcription Factor 2/genetics , Animals , Apoptosis , Carcinogens/toxicity , Cell Proliferation , Cyclin D1/metabolism , DNA/biosynthesis , Epidermis/drug effects , Epidermis/metabolism , Epidermis/pathology , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/pathology , Mice , Mice, Knockout , Papilloma/chemically induced , Papilloma/pathology , Presenilin-1/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Receptor, Notch1/metabolism , Skin Neoplasms/chemically induced , Skin Neoplasms/pathology , Tetradecanoylphorbol Acetate/toxicity , Tissue Array Analysis , Tumor Suppressor Proteins/analysis , Tumor Suppressor Proteins/genetics , beta Catenin/metabolism
7.
J Immunol ; 181(7): 4523-33, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18802055

ABSTRACT

IL-23 p19/p40, produced by macrophages and dendritic cells, is critical for development of Th17 in several autoimmune diseases. In this study, bone marrow-derived (BMM) and splenic macrophages (SPM) from SJL/J mice, susceptible to autoimmune demyelinating disease following Theiler's virus (TMEV) infection, expressed IL-23 in response to TMEV. We identified potential binding sites for IFN response factor (IRF)-3 (nt -734 to -731), Sma- and Mad-related protein (SMAD)-3 (nt -584 to -581), activating transcription factor (ATF)-2 (nt -571 to -568), IRF-7 (nt -533 to-525), and NF-kappaB (nt -215 to -209) in the murine p19 promoter. The p19(prom) in the pGL3 promoter-reporter vector responded to TMEV or poly(I:C), a TLR3 agonist in the RAW264.7 macrophage cell line. Deletions upstream from the IRF-3 site and mutations at the IRF-3, SMAD-3, ATF-2, or NF-kappaB, but not the IRF-7, sites significantly reduced promoter activity. ATF-2 or SMAD-3, but not IRF-3, short-hairpin RNA reduced p19 promoter activity and protein expression in RAW264.7 cells responding to TMEV. Chromosomal DNA immunoprecipitation assays revealed that SMAD-3 and ATF-2 bind to the endogenous p19 promoter in RAW264.7 cells and SJL/J SPM following challenge with TMEV. TGF-beta1, which activates SMAD-3, was induced in RAW264.7 cells, BMM, and SPM by TMEV. Neutralizing Ab to TGF-beta1 eliminated TMEV-induced IL-23 production and SMAD-3 activation in RAW264.7 cells, BMM, and SPM. Activation of ATF-2 was JNK, but not p38 or ERK MAPK dependent. Inhibition of the JNK, but also the ERK MAPK pathways decreased expression of p19. These results suggest that ATF-2 and SMAD-3 are transcription factors, which are, in addition to NF-kappaB, essential for IL-23 p19 expression.


Subject(s)
Activating Transcription Factor 2/genetics , Interleukin-23 Subunit p19/biosynthesis , Interleukin-23 Subunit p19/genetics , Macrophages/immunology , Macrophages/metabolism , Promoter Regions, Genetic/immunology , Smad3 Protein/genetics , Activating Transcription Factor 2/metabolism , Activating Transcription Factor 2/physiology , Animals , Base Sequence , Cell Line , Female , Gene Expression Regulation, Viral/immunology , Humans , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/physiology , Interferon Regulatory Factor-7/metabolism , Interleukin-23 Subunit p19/metabolism , Macrophages/virology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Mice , Molecular Sequence Data , NF-kappa B/metabolism , NF-kappa B/physiology , Protein Binding/genetics , Protein Binding/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Smad3 Protein/metabolism , Smad3 Protein/physiology , Theilovirus/immunology , Toll-Like Receptor 3/deficiency , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/physiology , Toll-Like Receptor 7/deficiency , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/physiology
8.
Bioessays ; 30(4): 314-27, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18348191

ABSTRACT

Activating Transcription Factor-2 is a sequence-specific DNA-binding protein that belongs to the bZIP family of proteins and plays diverse roles in the mammalian cells. In response to stress stimuli, it activates a variety of gene targets including cyclin A, cyclin D and c-jun, which are involved in oncogenesis in various tissue types. ATF-2 expression has been correlated with maintenance of a cancer cell phenotype. However, other studies demonstrate an antiproliferative or apoptotic role for ATF-2. In this review, we summarize the signaling pathways that activate ATF-2, as well as its downstream targets. We examine the role of ATF-2 in carcinogenesis with respect to other bZIP proteins, using data from studies in human cancer cell lines, human tumours and mouse models, and we propose a potential model for its function in carcinogenesis, as well as a theoretical basis for its utility in anticancer drug design.


Subject(s)
Activating Transcription Factor 2/genetics , Activating Transcription Factor 2/physiology , Neoplasms/metabolism , Activating Transcription Factor 2/metabolism , Animals , Apoptosis , Cell Adhesion , Cell Line, Tumor , Cell Transformation, Neoplastic , DNA-Binding Proteins/metabolism , Dimerization , Humans , Mice , Models, Biological , Models, Genetic , Signal Transduction , Transcription Factors/metabolism
9.
J Biomed Biotechnol ; 2009: 726728, 2009.
Article in English | MEDLINE | ID: mdl-19390583

ABSTRACT

Our method concentrates on and constructs the distinguished single gene network. An integrated method was proposed based on linear programming and a decomposition procedure with integrated analysis of the significant function cluster using Kappa statistics and fuzzy heuristic clustering. We tested this method to identify ATF2 regulatory network module using data of 45 samples from the same GEO dataset. The results demonstrate the effectiveness of such integrated way in terms of developing novel prognostic markers and therapeutic targets.


Subject(s)
Fuzzy Logic , Gene Regulatory Networks , Activating Transcription Factor 2/genetics , Activating Transcription Factor 2/physiology , Cluster Analysis , Data Interpretation, Statistical , Databases, Genetic , Humans , Oligonucleotide Array Sequence Analysis
10.
Nucleic Acids Res ; 35(4): 1312-21, 2007.
Article in English | MEDLINE | ID: mdl-17267404

ABSTRACT

The transcriptional activation of CHOP (a CCAAT/enhancer-binding protein-related gene) by amino acid deprivation involves the activating transcription factor 2 (ATF2) and the activating transcription factor 4 (ATF4) binding the amino acid response element (AARE) within the promoter. Using a chromatin immunoprecipitation approach, we report that in vivo binding of phospho-ATF2 and ATF4 to CHOP AARE are associated with acetylation of histones H4 and H2B in response to amino acid starvation. A time course analysis reveals that ATF2 phosphorylation precedes histone acetylation, ATF4 binding and the increase in CHOP mRNA. We also show that ATF4 binding and histone acetylation are two independent events that are required for the CHOP induction upon amino acid starvation. Using ATF2-deficient mouse embryonic fibroblasts, we demonstrate that ATF2 is essential in the acetylation of histone H4 and H2B in vivo. The role of ATF2 on histone H4 acetylation is dependent on its binding to the AARE and can be extended to other amino acid regulated genes. Thus, ATF2 is involved in promoting the modification of the chromatin structure to enhance the transcription of a number of amino acid-regulated genes.


Subject(s)
Activating Transcription Factor 2/physiology , Amino Acids/metabolism , Histones/metabolism , Transcription Factor CHOP/genetics , Transcriptional Activation , Acetylation , Activating Transcription Factor 4/metabolism , Animals , Cells, Cultured , Mice , Phosphorylation , Response Elements , Transcription Factor CHOP/biosynthesis
11.
Endocrinology ; 149(12): 6311-25, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18719024

ABSTRACT

Stimulation of GnRH receptors enhances expression of activating transcription factor (ATF) 3 in a pituitary gonadotroph cell line. The signaling pathway requires elevated cytosolic Ca2+ levels and activation of ERK and c-Jun N-terminal protein kinase. The signaling cascade was blocked by overexpression of either MAPK phosphatase (MKP)-1 or MAPK phosphatase-5 that dephosphorylate nuclear ERK and c-Jun N-terminal protein kinase. In addition, ATF3 biosynthesis was impaired after lentiviral-mediated expression of a constitutively active mutant of calcineurin A. Thus, MKP-1, MKP-5, and calcineurin may function as shut-off devices for GnRH receptor signaling. Expression of dominant-negative mutants of early growth response protein (Egr)-1, cAMP response element binding protein (CREB), and ATF2 blocked the biosynthesis of ATF3, indicating that these transcription factors connect the intracellular signaling cascade elicited by activation of GnRH receptors with transcription of the ATF3 gene. This view was corroborated by chromatin immunoprecipitation experiments revealing that Egr-1 and the phosphorylated forms of CREB and ATF2 bound to the 5'-upstream region of the ATF3 gene in buserelin-stimulated gonadotrophs. Together the data indicate that the ATF3 gene is a bona fide target gene of Egr-1, CREB, and ATF2 in gonadotrophs. Moreover, we show that in gonadotrophs ATF3 bound to its own promoter under physiological conditions. The analysis of a lentiviral-transmitted ATF3 promoter/luciferase reporter gene, embedded into the chromatin of the cells, revealed that ATF3 blocked the activity of its own promoter. We additionally identified the chromogranin B gene as bona fide target gene of ATF3 in gonadotrophs.


Subject(s)
Activating Transcription Factor 2/physiology , Activating Transcription Factor 3/biosynthesis , Cyclic AMP Response Element-Binding Protein/physiology , Early Growth Response Protein 1/physiology , Gonadotrophs/metabolism , Receptors, LHRH/physiology , Activating Transcription Factor 2/genetics , Activating Transcription Factor 2/metabolism , Activating Transcription Factor 3/genetics , Animals , Base Sequence , Blotting, Western , Buserelin/pharmacology , Calcineurin/genetics , Calcineurin/metabolism , Calcium/metabolism , Cell Line , Chromatin Immunoprecipitation , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Gonadotrophs/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , Kinetics , Mice , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Mutation , Phosphorylation/drug effects , Protein Binding/drug effects , Transfection
12.
Gene ; 421(1-2): 81-8, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18577431

ABSTRACT

The protease angiotensin converting enzyme (ACE) is a key regulator of blood pressure homeostasis, and is responsible for proteolytic activation of angiotensin I to angiotensin II (Ang II), a potent vasoconstrictor, and proteolytic inactivation of bradykinin, a vasodilator. Recent studies have also implicated ACE and Ang II dysregulation in the progression of fibrotic tissue diseases. Although many studies have utilized bovine tissues and cells for investigating the regulation of ACE gene expression, the bovine ACE promoter has not been previously characterized. Here we present the analysis of the bovine ACE promoter. We investigated cis elements regulated by phorbol 12-myristate 13-acetate (PMA). Sequence analysis shows that the bovine ACE promoter contains several putative binding sites for the transcription factors ATF-2, Ets-1, Egr-1 and SP1/SP3. Chromatin immunoprecipitation (ChIP) indicated that the activation of the bovine ACE promoter by PMA involves histone H4 acetylation, and that PMA induced Egr-1 and ATF-2 binding to the ACE promoter, whereas Ets-1 binding was suppressed by PMA. The regulatory roles of these transcription factors in the bovine ACE gene regulation were confirmed by co-expression of either wild type or dominant negative transcription factors with the luciferase reporter constructs. The bovine and human ACE promoters share similarities in binding sites for transcription factors and PMA regulation within the core regions but contain significant differences in the distal promoter regions.


Subject(s)
Activating Transcription Factor 2/physiology , Early Growth Response Protein 1/physiology , Peptidyl-Dipeptidase A/genetics , Promoter Regions, Genetic , Proto-Oncogene Protein c-ets-1/physiology , 5' Flanking Region , Amino Acid Sequence , Animals , Base Sequence , Cattle , Chromatin Immunoprecipitation , Cloning, Molecular , Gene Expression Regulation , Molecular Sequence Data , Peptidyl-Dipeptidase A/chemistry , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic
13.
Fungal Genet Biol ; 45(2): 103-16, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17625933

ABSTRACT

A defect in mcb, encoding the cAMP-dependent protein kinase A (PKA) regulatory subunit in Neurospora crassa, which confers an apolar growth phenotype, is accompanied by an increase in PKA activity levels. Both PKA and CRE-1 [a key carbon catabolite repression (CCR) regulator] mediate the cellular response to carbon-source availability. Inactivation of the cre-1 gene resulted in reduced growth rate, abnormal hyphal morphology and altered CCR. Both PKA and CRE-1 affected morphology in a carbon-dependent manner, as fructose suppressed the apolar morphology of the mcb strain and enabled faster growth of the Deltacre-1 mutant. An increase in cre-1 transcript abundance was observed in mcb and a reduction in PKA activity levels was measured in Deltacre-1. CRE-1 is involved in determining PKA-dependent polarity, as an mcb;Deltacre-1 strain displayed partial reestablishment of hyphal polarity. Taken together, our results demonstrate regulatory interactions between PKA and CRE-1 that affect cell polarity in a filamentous fungus.


Subject(s)
Activating Transcription Factor 2/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , Fungal Proteins/physiology , Gene Expression Regulation, Fungal , Neurospora crassa/physiology , Cell Polarity , Fructose , Genes, Fungal , Hyphae , Protein Subunits/genetics
14.
Exp Mol Med ; 40(6): 699-708, 2008 Dec 31.
Article in English | MEDLINE | ID: mdl-19116455

ABSTRACT

Expression of protein kinase C-delta (PKCdelta) is up-regulated by apoptosis-inducing stimuli. However, very little is known about the signaling pathways that control PKCdelta gene transcription. In the present study, we demonstrate that JNK stimulates PKCdelta gene expression via c-Jun and ATF2 in response to the anticancer agent doxorubicin (DXR) in mouse lymphocytic leukemia L1210 cells. Luciferase reporter assays showed that DXR-induced activation of the PKCdelta promoter was enhanced by ectopic expression of JNK1, c-Jun, or ATF2, whereas it was strongly reduced by expression of dominant negative JNK1 or by treatment with the JNK inhibitor SP600125. Furthermore, point mutations in the core sequence of the c-Jun/ATF2 binding site suppressed DXR-induced activation of the PKCdelta promoter. Our results suggest an additional role for a JNK signaling cascade in DXR-induced PKCdelta gene expression.


Subject(s)
Activating Transcription Factor 2/physiology , Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Mitogen-Activated Protein Kinase 8/physiology , Protein Kinase C-delta/metabolism , Proto-Oncogene Proteins c-jun/physiology , Animals , Anthracenes/pharmacology , Apoptosis , Cell Line, Tumor , Mice , Mutation , Promoter Regions, Genetic , Protein Kinase C-delta/genetics , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , Signal Transduction/physiology , Transcription, Genetic
15.
Cancer Res ; 66(21): 10487-96, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-17079470

ABSTRACT

Mounting evidence suggests a role for matrix metalloproteinase (MMP)-2 in the malignant progression of breast cancer cells. We showed previously that H-Ras, but not N-Ras, induced invasion of MCF10A human breast epithelial cells through Rac-MKK3/6-p38 pathway resulted in MMP-2 up-regulation. Activation of p38 pathway by MKK6 caused a selective up-regulation of MMP-2. In this study, we aimed to elucidate the transcriptional regulation of MMP-2 by p38 pathway leading to the invasive phenotype of MCF10A cells. By using 5' deletion mutant constructs of MMP-2 promoter, we showed that deletion of the region containing activator protein-1 (AP-1) site caused the greatest reduction of MMP-2 promoter activity both in MKK6- and H-Ras-activated MCF10A cells, suggesting that the AP-1 binding site is critical for the MMP-2 promoter activation. DNA binding and transcriptional activities of AP-1 were increased by MKK6 or H-Ras as evidenced by electrophoretic mobility shift assay and luciferase assay using an AP-1-driven plasmid. By doing immunoinhibition assay and chromatin immunoprecipitation assay, we revealed the activating transcription factor (ATF) 2 as a transcription factor for MMP-2 gene expression through binding to the functional AP-1 site. Activation of ATF2, which depended on p38 activity, was crucial for MMP-2 promoter activity as well as induction of invasive and migrative phenotypes in MCF10A cells. This is the first report revealing ATF2 as an essential transcription factor linking MKK3/6-p38 signaling pathway to MMP-2 up-regulation, providing evidence for a direct role of ATF2 activation in malignant phenotypic changes of human breast epithelial cells.


Subject(s)
Activating Transcription Factor 2/physiology , Breast Neoplasms/pathology , Breast/metabolism , Matrix Metalloproteinase 2/genetics , Transcriptional Activation , p38 Mitogen-Activated Protein Kinases/physiology , Binding Sites , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Epithelial Cells/metabolism , Female , Genes, ras/physiology , Humans , MAP Kinase Kinase 6/physiology , Neoplasm Invasiveness , Transcription Factor AP-1/metabolism
16.
Diabetes ; 55(8): 2301-10, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16873694

ABSTRACT

In metabolic syndrome, a systemic deregulation of the insulin pathway leads to a combined deregulation of insulin-regulated metabolism and cardiovascular functions. Free fatty acids (FFAs), which are increased in metabolic syndrome, inhibit insulin signaling and induce metabolic insulin resistance. This study was designed to examine FFAs' effects on vascular insulin signaling and endothelial nitric oxide (NO) synthase (eNOS) activation in endothelial cells. We showed that FFAs inhibited insulin signaling and eNOS activation through different mechanisms. While linoleic acid inhibited Akt-mediated eNOS phosphorylation, palmitic acid appeared to affect the upstream signaling. Upregulation of PTEN (phosphatase and tensin homolog deleted on chromosome 10) activity and transcription by palmitic acid mediated the inhibitory effects on insulin signaling. We further found that activated stress signaling p38, but not Jun NH(2)-terminal kinase, was involved in PTEN upregulation. The p38 target transcriptional factor activating transcription factor (ATF)-2 bound to the PTEN promoter, which was increased by palmitic acid treatment. In summary, both palmitic acid and linoleic acid exert inhibitory effect on insulin signaling and eNOS activation in endothelial cells. Palmitic acid inhibits insulin signaling by promoting PTEN activity and its transcription through p38 and its downstream transcription factor ATF-2. Our findings suggest that FFA-mediated inhibition of vascular insulin signaling and eNOS activation may contribute to cardiovascular diseases in metabolic syndrome.


Subject(s)
Endothelial Cells/drug effects , Fatty Acids, Nonesterified/pharmacology , Insulin/metabolism , Nitric Oxide Synthase Type III/metabolism , PTEN Phosphohydrolase/genetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Activating Transcription Factor 2/physiology , Cells, Cultured , Endothelial Cells/physiology , Enzyme Activation/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Linoleic Acid/pharmacology , PTEN Phosphohydrolase/physiology , Palmitic Acid/pharmacology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/analysis , Signal Transduction/drug effects , Signal Transduction/physiology , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
18.
J Invest Dermatol ; 137(2): 466-474, 2017 02.
Article in English | MEDLINE | ID: mdl-27729279

ABSTRACT

The phototherapeutic effects of visible red light on skin have been extensively investigated, but the underlying biological mechanisms remain poorly understood. We aimed to elucidate the protective mechanism of visible red light in terms of DNA repair of UV-induced oxidative damage in normal human dermal fibroblasts. The protective effect of visible red light on UV-induced DNA damage was identified by several assays in both two-dimensional and three-dimensional cell culture systems. With regard to the protective mechanism of visible red light, our data showed alterations in base excision repair mediated by growth arrest and DNA damage inducible, alpha (GADD45A). We also observed an enhancement of the physical activity of GADD45A and apurinic/apyrimidinic endonuclease 1 (APE1) by visible red light. Moreover, UV-induced DNA damages were diminished by visible red light in an APE1-dependent manner. On the basis of the decrease in GADD45A-APE1 interaction in the activating transcription factor-2 (ATF2)-knockdown system, we suggest a role for ATF2 modulation in GADD45A-mediated DNA repair upon visible red light exposure. Thus, the enhancement of GADD45A-mediated base excision repair modulated by ATF2 might be a potential protective mechanism of visible red light.


Subject(s)
Cell Cycle Proteins/physiology , Cytoprotection , DNA Repair , Light , Nuclear Proteins/physiology , Skin/radiation effects , Activating Transcription Factor 2/physiology , Cells, Cultured , DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , Fibroblasts/radiation effects , Humans , Skin/metabolism , Tumor Suppressor Protein p53/physiology
19.
Cancer Chemother Pharmacol ; 79(2): 369-380, 2017 02.
Article in English | MEDLINE | ID: mdl-28120035

ABSTRACT

PURPOSE: Nrf2 and its role in controlling levels of the AKR family of aldo-keto reductases which have been implicated in resistance to platinum-based chemotherapy was studied in ovarian, cervical and lung cell lines. METHODS: Nrf2 shRNA knockdowns of cells from different tumor origins were prepared to determine the role of this factor in producing resistance to platinum chemotherapy. RESULTS: Nrf2 knockdowns resulted in marked decreases in AKR1C1, AKR1C2 and to a lesser extent AKR1C3. Additionally, all other candidate enzymes GSTπ and TRX1 were decreased, but their role was difficult to correlate to cytotoxicity. Nrf2 knockdowns exhibited marked increases in mitochondrial membrane depolarization and ROS production following cisplatin treatment, with the cervical ME180R knockdowns exhibiting the greatest effect (AKR1C1 and AKR1C2 levels were decreased in the ME180R and SKOV3 cells to near zero). Oxaliplatin tended to parallel cisplatin, except it markedly stimulated O2- production not [Formula: see text] by oxaliplatin treatment of the ME180R cells. The pJNK/p38 pathway has been implicated in cisplatin cytotoxicity, and significant phosphorylation of pJNK was observed in the SKOV3 and ME180R and p38 in the SKOV3 knockdowns. Phosphorylation of ATF2 was decreased in the Nrf2 knockdowns (Crf38, Srf6, Arf5) which could affect its interaction with JNK and p38. Oxaliplatin treatment showed minimal effects on the JNK/p38 pathway, showing that its mode of action is different although ROS generation appeared an initial step with both drugs. CONCLUSIONS: Nrf2 controls a multitude of different candidate genes; however, it did markedly modulate cisplatin resistance through the AKR family. This involved ROS production and activation of the pJNK/p38 pathway with involvement of ATF2.


Subject(s)
Activating Transcription Factor 2/physiology , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , NF-E2-Related Factor 2/physiology , Organoplatinum Compounds/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Oxaliplatin , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Neurotoxicology ; 27(2): 158-63, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16185767

ABSTRACT

Several human diseases have been associated with the overproduction of reactive oxygen species (ROS) and subsequently various antioxidants emerged as potential therapeutic agents that scavenge ROS. As an oxidative stress model of human disease, we used hydrogen peroxide (H2O2) to study effect of ROS on C6 glioma cells as a surrogate for astrocytes. H2O2 induced dose- and time-dependent apoptotic cell death which was preceded by growth arrest, and transiently activated the signalling proteins ATF-2, ERK1/2 and AKT in C6 glioma cells. While several antioxidants failed to block H2O2-induced apoptosis of these cells, Ginkgo biloba extract (EGb) totally prevented the cell death and growth inhibition induced by H2O2. Interestingly, EGb did not prevent the activation of ATF-2, ERK1/2 and AKT induced by H2O2 excluding the role of these factors in the pro-apoptotic effect of H2O2. We have previously shown that the lipid-lowering drug, simvastatin, causes apoptotic cell death in C6 glioma cells [Koyuturk M, Ersoz M, Altiok N. Simvastatin induces proliferation inhibition and apoptosis in C6 glioma cells via c-jun N-terminal kinase. Neurosci Lett 2004;370(2-3):212-7]. However, in parallel experiments with H2O2, EGb was unable to prevent cell death induced by simvastatin suggesting the involvement of separate signalling pathways between H2O2 and simvastatin. Thus, EGb and other plant flavonoids might have potential as protective agents against apoptosis through scavenging ROS upon cerebral or myocardial diseases associated with free radical generation.


Subject(s)
Cell Death/drug effects , Ginkgo biloba/chemistry , Hydrogen Peroxide/toxicity , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Neuroglia/drug effects , Oxidants/toxicity , Simvastatin/toxicity , Activating Transcription Factor 2/physiology , Apoptosis/drug effects , Blotting, Western , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Extracellular Signal-Regulated MAP Kinases/physiology , Glioma/pathology , Humans , Immunohistochemistry , Immunoprecipitation , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL