Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Curr Opin Genet Dev ; 11(5): 497-504, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11532390

ABSTRACT

Skeletal muscle cells have provided an especially auspicious system in which to dissect the roles of chromatin structure in the control of cell growth, differentiation, and development. The MyoD and MEF2 families of transcription factors act cooperatively to regulate the expression of skeletal muscle-specific genes. Recent studies have shown that these two classes of transcription factors associate with histone acetyltransferases and histone deacetylases to control the activation and repression, respectively, of the muscle differentiation program. Signaling systems that regulate the growth and differentiation of muscle cells act, at least in part, by regulating the intracellular localization and associations of these chromatin remodeling enzymes with myogenic transcription factors. We describe the molecules and mechanisms involved in chromatin remodeling during skeletal muscle development.


Subject(s)
Acetyltransferases/genetics , Acetyltransferases/metabolism , Histone Deacetylases/metabolism , Muscle, Skeletal/enzymology , Muscle, Skeletal/growth & development , Saccharomyces cerevisiae Proteins , Active Transport, Cell Nucleus , Animals , Carrier Proteins , Cell Cycle Proteins/metabolism , Chromatin/enzymology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Histone Acetyltransferases , Humans , MEF2 Transcription Factors , Muscle Development/genetics , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , MyoD Protein/genetics , MyoD Protein/metabolism , Myogenic Regulatory Factors , Nuclear Receptor Co-Repressor 2 , Nuclear Receptor Coactivator 2 , Phosphoproteins/metabolism , Repressor Proteins/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , p300-CBP Transcription Factors
2.
Mol Cell Biol ; 21(18): 6312-21, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11509672

ABSTRACT

Activation of muscle-specific genes by the MEF2 transcription factor is inhibited by class II histone deacetylases (HDACs) 4 and 5, which contain carboxy-terminal deacetylase domains and amino-terminal extensions required for association with MEF2. The inhibitory action of HDACs is overcome by myogenic signals which disrupt MEF2-HDAC interactions and stimulate nuclear export of these transcriptional repressors. Nucleocytoplasmic trafficking of HDAC5 is mediated by binding of the chaperone protein 14-3-3 to two phosphoserine residues (Ser-259 and Ser-498) in its amino-terminal extension. Here we show that HDAC4 and -5 each contain a signal-responsive nuclear export sequence (NES) at their extreme carboxy termini. The NES is conserved in another class II HDAC, HDAC7, but is absent in class I HDACs and the HDAC-related corepressor, MEF2-interacting transcription repressor. Our results suggest that this conserved NES is inactive in unphosphorylated HDAC5, which is localized to the nucleus, and that calcium-calmodulin-dependent protein kinase (CaMK)-dependent binding of 14-3-3 to phosphoserines 259 and 498 activates the NES, with consequent export of the transcriptional repressor to the cytoplasm. A single amino acid substitution in this NES is sufficient to retain HDAC5 in the nucleus in the face of CaMK signaling. These findings provide molecular insight into the mechanism by which extracellular cues alter chromatin structure to promote muscle differentiation and other MEF2-regulated processes.


Subject(s)
Histone Deacetylases/genetics , Repressor Proteins , Amino Acid Sequence , Animals , COS Cells , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Histone Deacetylase 2 , Histone Deacetylases/metabolism , MEF2 Transcription Factors , Molecular Sequence Data , Myogenic Regulatory Factors , Sequence Deletion , Signal Transduction/genetics , Structure-Activity Relationship , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Proc Natl Acad Sci U S A ; 98(13): 7354-9, 2001 Jun 19.
Article in English | MEDLINE | ID: mdl-11390982

ABSTRACT

Activation of muscle-specific genes by members of the myocyte enhancer factor 2 (MEF2) and MyoD families of transcription factors is coupled to histone acetylation and is inhibited by class II histone deacetylases (HDACs) 4 and 5, which interact with MEF2. The ability of HDAC4 and -5 to inhibit MEF2 is blocked by phosphorylation of these HDACs at two conserved serine residues, which creates docking sites for the intracellular chaperone protein 14-3-3. When bound to 14-3-3, HDACs are released from MEF2 and transported to the cytoplasm, thereby allowing MEF2 to stimulate muscle-specific gene expression. MEF2-interacting transcription repressor (MITR) shares homology with the amino-terminal regions of HDAC4 and -5, but lacks an HDAC catalytic domain. Despite the absence of intrinsic HDAC activity, MITR acts as a potent inhibitor of MEF2-dependent transcription. Paradoxically, however, MITR has minimal inhibitory effects on the skeletal muscle differentiation program. We show that a substitution mutant of MITR containing alanine in place of two serine residues, Ser-218 and Ser-448, acts as a potent repressor of myogenesis. Our findings indicate that promyogenic signals antagonize the inhibitory action of MITR by targeting these serines for phosphorylation. Phosphorylation of Ser-218 and Ser-448 stimulates binding of 14-3-3 to MITR, disrupts MEF2:MITR interactions, and alters the nuclear distribution of MITR. These results reveal a role for MITR as a signal-dependent regulator of muscle differentiation.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Embryonic and Fetal Development , Gene Expression Regulation, Developmental , Muscle, Skeletal/physiology , 14-3-3 Proteins , Animals , COS Cells , Cell Differentiation , Cell Line , Chlorocebus aethiops , DNA-Binding Proteins/metabolism , Histone Deacetylases/metabolism , MEF2 Transcription Factors , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/embryology , MyoD Protein/metabolism , Myogenic Regulatory Factors , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Transcription, Genetic , Transfection , Tyrosine 3-Monooxygenase/metabolism
4.
Proc Natl Acad Sci U S A ; 98(6): 3328-33, 2001 Mar 13.
Article in English | MEDLINE | ID: mdl-11248078

ABSTRACT

Signaling events controlled by calcineurin promote cardiac hypertrophy, but the degree to which such pathways are required to transduce the effects of various hypertrophic stimuli remains uncertain. In particular, the administration of immunosuppressive drugs that inhibit calcineurin has inconsistent effects in blocking cardiac hypertrophy in various animal models. As an alternative approach to inhibiting calcineurin in the hearts of intact animals, transgenic mice were engineered to overexpress a human cDNA encoding the calcineurin-binding protein, myocyte-enriched calcineurin-interacting protein-1 (hMCIP1) under control of the cardiac-specific, alpha-myosin heavy chain promoter (alpha-MHC). In unstressed mice, forced expression of hMCIP1 resulted in a 5-10% decline in cardiac mass relative to wild-type littermates, but otherwise produced no apparent structural or functional abnormalities. However, cardiac-specific expression of hMCIP1 inhibited cardiac hypertrophy, reinduction of fetal gene expression, and progression to dilated cardiomyopathy that otherwise result from expression of a constitutively active form of calcineurin. Expression of the hMCIP1 transgene also inhibited hypertrophic responses to beta-adrenergic receptor stimulation or exercise training. These results demonstrate that levels of hMCIP1 producing no apparent deleterious effects in cells of the normal heart are sufficient to inhibit several forms of cardiac hypertrophy, and suggest an important role for calcineurin signaling in diverse forms of cardiac hypertrophy. The future development of measures to increase expression or activity of MCIP proteins selectively within the heart may have clinical value for prevention of heart failure.


Subject(s)
Calcineurin Inhibitors , Cardiomyopathy, Dilated/prevention & control , Muscle Proteins/physiology , Animals , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , DNA-Binding Proteins , Female , Gene Expression , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Genetic , Muscle Proteins/genetics , Myosin Heavy Chains/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology
5.
J Biol Chem ; 276(1): 35-9, 2001 Jan 05.
Article in English | MEDLINE | ID: mdl-11022042

ABSTRACT

The class II histone deacetylases (HDACs) 4, 5, and 7 share a common structural organization, with a carboxyl-terminal catalytic domain and an amino-terminal extension that mediates interactions with members of the myocyte enhancer factor-2 (MEF2) family of transcription factors. Association of these HDACs with MEF2 factors represses transcription of MEF2 target genes. MEF2-interacting transcription repressor (MITR) shares homology with the amino-terminal extensions of class II HDACs and also acts as a transcriptional repressor, but lacks a histone deacetylase catalytic domain. This suggests that MITR represses transcription by recruiting other corepressors. We show that the amino-terminal regions of MITR and class II HDACs interact with the transcriptional corepressor, COOH-terminal-binding protein (CtBP), through a CtBP-binding motif (P-X-D-L-R) conserved in MITR and HDACs 4, 5, and 7. Mutation of this sequence in MITR abolishes interaction with CtBP and impairs, but does not eliminate, the ability of MITR to inhibit MEF2-dependent transcription. The residual repressive activity of MITR mutants that fail to bind CtBP can be attributed to association with other HDAC family members. These findings reveal CtBP-dependent and -independent mechanisms for transcriptional repression by MITR and show that MITR represses MEF2 activity through recruitment of multicomponent corepressor complexes that include CtBP and HDACs.


Subject(s)
Carrier Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Phosphoproteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/genetics , Alcohol Oxidoreductases , Amino Acid Motifs , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Line , DNA-Binding Proteins/chemistry , Histone Deacetylases/chemistry , Histone Deacetylases/classification , Histone Deacetylases/metabolism , MEF2 Transcription Factors , Macromolecular Substances , Mice , Molecular Sequence Data , Mutation , Myogenic Regulatory Factors , Phosphoproteins/chemistry , Phosphoproteins/genetics , Precipitin Tests , Protein Binding , Repressor Proteins/chemistry , Repressor Proteins/genetics , Transcription Factors/metabolism , Two-Hybrid System Techniques
6.
Proc Natl Acad Sci U S A ; 97(26): 14400-5, 2000 Dec 19.
Article in English | MEDLINE | ID: mdl-11114197

ABSTRACT

Skeletal muscle differentiation is controlled by interactions between myocyte enhancer factor-2 (MEF2) and myogenic basic helix-loop-helix transcription factors. Association of MEF2 with histone deacetylases (HDAC) -4 and -5 results in repression of MEF2 target genes and inhibition of myogenesis. Calcium/calmodulin-dependent protein kinase (CaMK) signaling promotes myogenesis by disrupting MEF2-HDAC complexes and stimulating HDAC nuclear export. To further define the mechanisms that confer CaMK responsiveness to HDAC4 and -5, we performed yeast two-hybrid screens to identify HDAC-interacting factors. These screens revealed interactions between HDAC4 and members of the 14-3-3 family of proteins, which function as signal-dependent intracellular chaperones. HDAC4 binds constitutively to 14-3-3 in yeast and mammalian cells, whereas HDAC5 binding to 14-3-3 is largely dependent on CaMK signaling. CaMK phosphorylates serines -259 and -498 in HDAC5, which subsequently serve as docking sites for 14-3-3. Our studies suggest that 14-3-3 binding to HDAC5 is required for CaMK-dependent disruption of MEF2-HDAC complexes and nuclear export of HDAC5, and implicate 14-3-3 as a signal-dependent regulator of muscle cell differentiation.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , DNA-Binding Proteins/metabolism , Histone Deacetylases/metabolism , Transcription Factors/metabolism , Tyrosine 3-Monooxygenase/metabolism , 14-3-3 Proteins , Animals , Binding Sites , Biological Transport , COS Cells , Cell Nucleus/metabolism , Chlorocebus aethiops , Histone Deacetylases/genetics , MEF2 Transcription Factors , Mice , Myogenic Regulatory Factors , Repressor Proteins/genetics , Repressor Proteins/metabolism , Saccharomyces cerevisiae , Serine/metabolism , Signal Transduction , Tyrosine 3-Monooxygenase/genetics
7.
Nat Med ; 6(11): 1221-7, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11062532

ABSTRACT

Calcium is central in the regulation of cardiac contractility, growth and gene expression. Variations in the amplitude, frequency and compartmentalization of calcium signals are decoded by calcium/calmodulin-dependent enzymes, ion channels and transcription factors. Understanding the circuitry for calcium signaling creates opportunities for pharmacological modification of cardiac function.


Subject(s)
Calcium Signaling/physiology , Heart/physiology , Myocardial Contraction/physiology , Animals , Calmodulin/physiology , Cardiomegaly/physiopathology , Gene Expression Regulation , Heart/growth & development , Humans
8.
Nature ; 408(6808): 106-11, 2000 Nov 02.
Article in English | MEDLINE | ID: mdl-11081517

ABSTRACT

Members of the myocyte enhancer factor-2 (MEF2) family of transcription factors associate with myogenic basic helix-loop-helix transcription factors such as MyoD to activate skeletal myogenesis. MEF2 proteins also interact with the class II histone deacetylases HDAC4 and HDAC5, resulting in repression of MEF2-dependent genes. Execution of the muscle differentiation program requires release of MEF2 from repression by HDACs, which are expressed constitutively in myoblasts and myotubes. Here we show that HDAC5 shuttles from the nucleus to the cytoplasm when myoblasts are triggered to differentiate. Calcium/calmodulin-dependent protein kinase (CaMK) signalling, which stimulates myogenesis and prevents formation of MEF2-HDAC complexes, also induces nuclear export of HDAC4 and HDAC5 by phosphorylation of these transcriptional repressors. An HDAC5 mutant lacking two CaMK phosphorylation sites is resistant to CaMK-mediated nuclear export and acts as a dominant inhibitor of skeletal myogenesis, whereas a cytoplasmic HDAC5 mutant is unable to block efficiently the muscle differentiation program. Our results highlight a mechanism for transcriptional regulation through signal- and differentiation-dependent nuclear export of a chromatin-remodelling enzyme, and suggest that nucleo-cytoplasmic trafficking of HDACs is involved in the control of cellular differentiation.


Subject(s)
Histone Deacetylases/metabolism , Muscle, Skeletal/cytology , Signal Transduction , Animals , COS Cells , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Differentiation , Cell Line , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Histone Deacetylases/genetics , Histones/metabolism , MEF2 Transcription Factors , Mutagenesis , Myogenic Regulatory Factors , Phosphorylation , Protein Transport , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/metabolism
9.
Mol Cell ; 6(2): 233-44, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10983972

ABSTRACT

Skeletal muscle differentiation is controlled by associations between myogenic basic-helix-loop-helix and MEF2 transcription factors. We show that chromatin associated with muscle genes regulated by these transcription factors becomes acetylated during myogenesis and that class II histone deacetylases (HDACs), which interact with MEF2, specifically suppress myoblast differentiation. These HDACs do not interact directly with MyoD, yet they suppress its myogenic activity through association with MEF2. Elevating the level of MyoD can override the repression imposed by HDACs on muscle genes. HDAC-mediated repression of myogenesis also can be overcome by CaM kinase and insulin-like growth factor (IGF) signaling. These findings reveal central roles for HDACs in chromatin remodeling during myogenesis and as intranuclear targets for signaling pathways controlled by IGF and CaM kinase.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation , Histone Deacetylases/metabolism , Muscle, Skeletal/physiology , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 4 , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Differentiation , Cell Line , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , MEF2 Transcription Factors , Mice , Molecular Sequence Data , Muscle, Skeletal/cytology , MyoD Protein/genetics , MyoD Protein/metabolism , Myogenic Regulatory Factors , Rats , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction , Transcription Factors/chemistry , Transcription Factors/genetics , Transfection
10.
EMBO J ; 19(9): 1963-73, 2000 May 02.
Article in English | MEDLINE | ID: mdl-10790363

ABSTRACT

Different patterns of motor nerve activity drive distinctive programs of gene transcription in skeletal muscles, thereby establishing a high degree of metabolic and physiological specialization among myofiber subtypes. Recently, we proposed that the influence of motor nerve activity on skeletal muscle fiber type is transduced to the relevant genes by calcineurin, which controls the functional activity of NFAT (nuclear family of activated T cell) proteins. Here we demonstrate that calcineurin-dependent gene regulation in skeletal myocytes is mediated also by MEF2 transcription factors, and is integrated with additional calcium-regulated signaling inputs, specifically calmodulin-dependent protein kinase activity. In skeletal muscles of transgenic mice, both NFAT and MEF2 binding sites are necessary for properly regulated function of a slow fiber-specific enhancer, and either forced expression of activated calcineurin or motor nerve stimulation up-regulates a MEF2-dependent reporter gene. These results provide new insights into the molecular mechanisms by which specialized characteristics of skeletal myofiber subtypes are established and maintained.


Subject(s)
Calcineurin/metabolism , Calcium Signaling , Calcium/physiology , DNA-Binding Proteins/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Nuclear Proteins , Transcription Factors/metabolism , Animals , Base Sequence , Calcineurin/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 4 , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Electric Stimulation , Enhancer Elements, Genetic/genetics , MEF2 Transcription Factors , Mice , Mice, Transgenic , Motor Neurons/physiology , Muscle Fibers, Fast-Twitch/cytology , Muscle Fibers, Fast-Twitch/enzymology , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/enzymology , Muscle Fibers, Slow-Twitch/cytology , Muscle Fibers, Slow-Twitch/enzymology , Muscle Fibers, Slow-Twitch/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/enzymology , Muscle, Skeletal/innervation , Myogenic Regulatory Factors , NFATC Transcription Factors , Organ Specificity , Phosphorylation , Protein Binding , Transcription Factors/genetics , Transcription Factors/physiology , Transcriptional Activation
11.
J Clin Invest ; 105(10): 1395-406, 2000 May.
Article in English | MEDLINE | ID: mdl-10811847

ABSTRACT

Hypertrophic growth is an adaptive response of the heart to diverse pathological stimuli and is characterized by cardiomyocyte enlargement, sarcomere assembly, and activation of a fetal program of cardiac gene expression. A variety of Ca(2+)-dependent signal transduction pathways have been implicated in cardiac hypertrophy, but whether these pathways are independent or interdependent and whether there is specificity among them are unclear. Previously, we showed that activation of the Ca(2+)/calmodulin-dependent protein phosphatase calcineurin or its target transcription factor NFAT3 was sufficient to evoke myocardial hypertrophy in vivo. Here, we show that activated Ca(2+)/calmodulin-dependent protein kinases-I and -IV (CaMKI and CaMKIV) also induce hypertrophic responses in cardiomyocytes in vitro and that CaMKIV overexpressing mice develop cardiac hypertrophy with increased left ventricular end-diastolic diameter and decreased fractional shortening. Crossing this transgenic line with mice expressing a constitutively activated form of NFAT3 revealed synergy between these signaling pathways. We further show that CaMKIV activates the transcription factor MEF2 through a posttranslational mechanism in the hypertrophic heart in vivo. Activated calcineurin is a less efficient activator of MEF2-dependent transcription, suggesting that the calcineurin/NFAT and CaMK/MEF2 pathways act in parallel. These findings identify MEF2 as a downstream target for CaMK signaling in the hypertrophic heart and suggest that the CaMK and calcineurin pathways preferentially target different transcription factors to induce cardiac hypertrophy.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cardiomegaly/etiology , Cardiomegaly/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins , Transcription Factors/metabolism , Animals , Atrial Natriuretic Factor/genetics , Calcineurin/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 4 , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cardiomegaly/genetics , Gene Expression Regulation , Genes, Reporter , Humans , Luciferases/genetics , MEF2 Transcription Factors , Mice , Mice, Transgenic , Myocardium/metabolism , Myogenic Regulatory Factors , Myosin Heavy Chains/genetics , NFATC Transcription Factors , Promoter Regions, Genetic , Rats , Signal Transduction
12.
Proc Natl Acad Sci U S A ; 97(8): 4070-5, 2000 Apr 11.
Article in English | MEDLINE | ID: mdl-10737771

ABSTRACT

Myocyte enhancer factor-2 (MEF2) transcription factors control muscle-specific and growth factor-inducible genes. We show that hypertrophic growth of cardiomyocytes in response to phenylephrine and serum is accompanied by activation of MEF2 through a posttranslational mechanism mediated by calcium, calmodulin-dependent protein kinase (CaMK), and mitogen-activated protein kinase (MAPK) signaling. CaMK stimulates MEF2 activity by dissociating class II histone deacetylases (HDACs) from the DNA-binding domain. MAPKs, which activate MEF2 by phosphorylation of the transcription activation domain, maximally stimulate MEF2 activity only when repression by HDACs is relieved by CaMK signaling to the DNA-binding domain. These findings identify MEF2 as an endpoint for hypertrophic stimuli in cardiomyocytes and demonstrate that MEF2 mediates synergistic transcriptional responses to the CaMK and MAPK signaling pathways by signal-dependent dissociation from HDACs.


Subject(s)
DNA-Binding Proteins/metabolism , Histone Deacetylases/metabolism , Signal Transduction , Transcription Factors/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , MEF2 Transcription Factors , Mice , Myogenic Regulatory Factors , Rats , Transcription, Genetic
13.
Oncogene ; 19(7): 841-9, 2000 Feb 17.
Article in English | MEDLINE | ID: mdl-10702792

ABSTRACT

Oncogenic mutations in ras lead to constitutive activation of downstream signaling pathways that modulate the activities of transcription factors. In turn, these factors control the expression of a subset of genes responsible for neoplastic cell transformation. Recent studies suggest that transcription factor NF-kappa B contributes to cell transformation by inhibiting the cell death signal activated by oncogenic Ras. In this study, inhibition of NF-kappa B activity by forced expression of a super-repressor form of I kappa B alpha, the major inhibitor of NF-kappa B, markedly decreased the growth rate, saturation density and tumorigenicity of oncogenic H-Ras transformed rat embryo fibroblasts. Such clonally isolated cells overexpressing I kappa B alpha super-repressor not only were viable but also exhibited no sign of spontaneous apoptosis. Inhibition of NF-kappa B in these cells was functionally demonstrated by both the loss of cytokine induced DNA binding activity and a profoundly increased sensitivity to cell death in response to TNF-alpha treatment. In contrast, inhibition of NF-kappa B activity in non-transformed fibroblasts had minimal effect on growth, but rendered the cells resistant to a subsequent transformation by H-ras oncogene. Similar results were also obtained with rat intestinal epithelial cells harboring an inducible ras oncogene. Taken together, these findings suggest that NF-kappa B activity is essential for abnormal cell proliferation and tumorigenicity activated by the ras oncogene and highlight an alternative functional role for NF-kappa B in oncogenic Ras-mediated cell transformation that is distinct from its anti-apoptotic activity. Oncogene (2000) 19, 841 - 849.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Genes, ras/physiology , NF-kappa B/physiology , Animals , Apoptosis/genetics , Cell Division/genetics , Cell Line, Transformed , Cell Survival/genetics , Cell Transformation, Neoplastic/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice , Mice, Nude , Rats
14.
Circ Res ; 87(12): E61-8, 2000 Dec 08.
Article in English | MEDLINE | ID: mdl-11110780

ABSTRACT

Calcineurin, a calcium/calmodulin-regulated protein phosphatase, modulates gene expression in cardiac and skeletal muscles during development and in remodeling responses such as cardiac hypertrophy that are evoked by environmental stresses or disease. Recently, we identified two genes encoding proteins (MCIP1 and MCIP2) that are enriched in striated muscles and that interact with calcineurin to inhibit its enzymatic activity. In the present study, we show that expression of MCIP1 is regulated by calcineurin activity in hearts of mice with cardiac hypertrophy, as well as in cultured skeletal myotubes. In contrast, expression of MCIP2 in the heart is not altered by activated calcineurin but responds to thyroid hormone, which has no effect on MCIP1. A approximately 900-bp intragenic segment located between exons 3 and 4 of the MCIP1 gene functions as an alternative promoter that responds to calcineurin. This region includes a dense cluster of 15 consensus binding sites for NF-AT transcription factors. Because MCIP proteins can inhibit calcineurin, these results suggest that MCIP1 participates in a negative feedback circuit to diminish potentially deleterious effects of unrestrained calcineurin activity in cardiac and skeletal myocytes. Inhibitory effects of MCIP2 on calcineurin activity may be pertinent to gene switching events driven by thyroid hormone in striated muscles. The full text of this article is available at http://www. circresaha.org.


Subject(s)
Calcineurin/physiology , Gene Expression Regulation , Muscle Proteins/genetics , Muscle, Skeletal/physiology , Proteins , Animals , Cells, Cultured , DNA-Binding Proteins , Exons , Humans , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL , Muscle Proteins/biosynthesis , RNA, Messenger/biosynthesis , Signal Transduction , Thyroid Hormones/physiology , Transcription, Genetic , Transfection
15.
Mol Immunol ; 37(12-13): 783-8, 2000.
Article in English | MEDLINE | ID: mdl-11275263

ABSTRACT

The immunoglobulin superfamily member CD83 is expressed on the surface of mature dendritic cells that present processed antigens to T lymphocytes. In addition, T cells acquire CD83 expression following mitogenic stimulation in vitro. Here we report two lines of evidence demonstrating that this inducible lymphocyte response is genetically programmed by transcription factor NF-kappaB and contingent upon proteolytic breakdown of its cytoplasmic inhibitor IkappaBalpha. First, signal-dependent induction of CD83 mRNA expression is blocked in both transformed and primary T cells harboring a degradation-resistant mutant of IkappaBalpha that constitutively represses NF-kappaB. Second, as revealed in gel retardation assays, the IkappaBalpha constitutive repressor prevents the inducible interaction of NF-kappaB with consensus recognition sites identified in the CD83 promoter. Given that IkappaBalpha is functionally coupled to the T-cell antigen receptor, these findings suggest that the downstream transcription unit for CD83 is triggered by NF-kappaB during an adaptive immune response.


Subject(s)
I-kappa B Proteins , Immunoglobulins/genetics , Lymphocyte Activation , Membrane Glycoproteins/genetics , NF-kappa B/metabolism , T-Lymphocytes/immunology , Antigens, CD , Binding Sites , DNA-Binding Proteins/genetics , Gene Expression Regulation , Genetic Engineering , Humans , Immunoglobulins/biosynthesis , Jurkat Cells , Membrane Glycoproteins/biosynthesis , NF-KappaB Inhibitor alpha , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/isolation & purification , Signal Transduction , CD83 Antigen
16.
Curr Opin Genet Dev ; 9(3): 267-74, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10377279

ABSTRACT

Cardiac hypertrophy is an adaptive response of the heart to a variety of intrinsic and extrinsic stimuli. The hypertrophic response, during which cardiomyocytes increase in size without undergoing cell division, initially serves to compensate for decreased cardiac output; however, prolonged hypertrophy can become detrimental, resulting in dilated cardiomyopathy and heart failure. Cardiac hypertrophy requires coupling of intracellular signal transduction systems with transcription factors that activate and maintain the hypertrophic program. Over the past year, signaling pathways involving G proteins, mitogen-activated protein kinases and calcium-responsive phosphatases have emerged as critical regulators of cardiac hypertrophy.


Subject(s)
Cardiomegaly/metabolism , Nuclear Proteins , Animals , Calcineurin/metabolism , Calcineurin Inhibitors , Cardiomegaly/genetics , Cardiomegaly/physiopathology , Cyclosporine/pharmacology , DNA-Binding Proteins/metabolism , Humans , MAP Kinase Signaling System , NFATC Transcription Factors , Transcription Factors/metabolism
17.
Mol Cell Biol ; 19(6): 4495-502, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10330188

ABSTRACT

Members of the GATA family of zinc finger transcription factors have been shown to play important roles in the control of gene expression in a variety of cell types. GATA-1, -2, and -3 are expressed primarily in hematopoietic cell lineages and are required for proliferation and differentiation of multiple hematopoietic cell types, whereas GATA-4, -5, and -6 are expressed in the heart, where they activate cardiac muscle structural genes. Friend of GATA-1 (FOG) is a multitype zinc finger protein that interacts with GATA-1 and serves as a cofactor for GATA-1-mediated transcription. FOG is coexpressed with GATA-1 in developing erythroid and megakaryocyte cell lineages and cooperates with GATA-1 to control erythropoiesis. We describe a novel FOG-related factor, FOG-2, that is expressed predominantly in the developing and adult heart, brain, and testis. FOG-2 interacts with GATA factors, and interaction of GATA-4 and FOG-2 results in either synergistic activation or repression of GATA-dependent cardiac promoters, depending on the specific promoter and the cell type in which they are tested. The properties of FOG-2 suggest its involvement in the control of cardiac and neural gene expression by GATA transcription factors.


Subject(s)
Brain/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Myocardium/metabolism , Amino Acid Sequence , Animals , COS Cells , Cells, Cultured , Cloning, Molecular , DNA-Binding Proteins/analysis , Databases, Factual , Embryo, Mammalian/anatomy & histology , Expressed Sequence Tags , GATA4 Transcription Factor , Humans , Immunohistochemistry , In Situ Hybridization , Male , Mice , Models, Genetic , Molecular Sequence Data , Promoter Regions, Genetic , Rats , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Testis/metabolism , Tissue Distribution , Transcription Factors/metabolism , Transcription, Genetic , Transfection , Zinc Fingers/genetics
18.
Circ Res ; 84(6): 735-40, 1999 Apr 02.
Article in English | MEDLINE | ID: mdl-10189362

ABSTRACT

Left ventricular hypertrophy (LVH) is a compensatory mechanism to cope with pressure overload. Recently, a calcineurin pathway mediating LVH and its prevention by cyclosporine was reported. We examined whether calcineurin mediates LVH due to pressure overload in mice. Pressure overload was induced by aortic banding in 53 mice (32 treated with cyclosporine [25 mg. kg-1. d-1], 21 treated with vehicle). There were 17 sham-operated mice (9 treated with vehicle, 8 treated with cyclosporine). At 3 weeks after surgery, LV weight to body weight was greater in the nontreatment banded group (4.39+/-0. 16 mg/g) than in the cyclosporine-treated banded group (3.95+/-0.14 mg/g, P<0.05), with both groups being greater compared with the entire group of sham-operated mice (3.02+/-0.04 mg/g). The pressure gradient between the ascending and abdominal aorta was not different between the cyclosporine-treated (49.6+/-6.1 mm Hg) and nontreatment groups (48.7+/-4.6 mm Hg). Although LV systolic pressure was lower in the cyclosporine-treated banded animals, LV systolic wall stress was similar in the nontreatment banded group and in the cyclosporine-treated group. However, LV dP/dt was lower (P=0.05) in the cyclosporine-treated banded group (4774+/-656 mm Hg/s) than in the nontreatment banded group (6604+/-516 mm Hg/s). During the protocol, 23 of 32 mice in the cyclosporine-treated group and 9 of 21 mice in the nontreatment group died. All deaths occurred within 10 days after surgery. Deaths caused by heart failure were 7.2-fold higher (P<0.05) in the cyclosporine-treated group, whereas deaths due to other causes were not different between the 2 groups. In addition, LV function of mice was assessed at 48 hours after banding; LV ejection fraction measured with echocardiography was lower (P<0.05) in the cyclosporine-treated banded group (66+/-3.0%) than in the nontreatment banded group (79+/-1.5%), whereas LV systolic wall stresses were similar. Calcineurin phosphatase activity was depressed similarly in both cyclosporine-treated groups compared with both nontreatment groups. Thus, cyclosporine could attenuate, but not prevent, LVH at the expense of inhibiting an important compensatory mechanism in response to pressure overload, resulting in reduced LV wall stress and function and increased susceptibility to decompensation and heart failure.


Subject(s)
Cyclosporine/pharmacology , Enzyme Inhibitors/pharmacology , Heart Failure/etiology , Hypertension/physiopathology , Hypertrophy, Left Ventricular/physiopathology , Animals , Aorta/physiology , Aorta, Abdominal/physiology , Aorta, Thoracic/physiology , Cyclosporine/blood , Disease Susceptibility , Enzyme Inhibitors/blood , Heart Failure/mortality , Heart Failure/physiopathology , Hypertension/mortality , Hypertrophy, Left Ventricular/mortality , Hypertrophy, Left Ventricular/pathology , Ligation , Male , Mice
19.
Proc Natl Acad Sci U S A ; 94(19): 10057-62, 1997 Sep 16.
Article in English | MEDLINE | ID: mdl-9294162

ABSTRACT

Members of the NF-kappaB/Rel and inhibitor of apoptosis (IAP) protein families have been implicated in signal transduction programs that prevent cell death elicited by the cytokine tumor necrosis factor alpha (TNF). Although NF-kappaB appears to stimulate the expression of specific protective genes, neither the identities of these genes nor the precise role of IAP proteins in this anti-apoptotic process are known. We demonstrate here that NF-kappaB is required for TNF-mediated induction of the gene encoding human c-IAP2. When overexpressed in mammalian cells, c-IAP2 activates NF-kappaB and suppresses TNF cytotoxicity. Both of these c-IAP2 activities are blocked in vivo by coexpressing a dominant form of IkappaB that is resistant to TNF-induced degradation. In contrast to wild-type c-IAP2, a mutant lacking the C-terminal RING domain inhibits NF-kappaB induction by TNF and enhances TNF killing. These findings suggest that c-IAP2 is critically involved in TNF signaling and exerts positive feedback control on NF-kappaB via an IkappaB targeting mechanism. Functional coupling of NF-kappaB and c-IAP2 during the TNF response may provide a signal amplification loop that promotes cell survival rather than death.


Subject(s)
Apoptosis/physiology , NF-kappa B/physiology , Tumor Necrosis Factor-alpha/physiology , Viral Proteins/physiology , Gene Expression Regulation/physiology , Humans , Inhibitor of Apoptosis Proteins , Jurkat Cells , Transcription, Genetic/physiology , Viral Proteins/genetics
20.
J Biol Chem ; 272(36): 22377-80, 1997 Sep 05.
Article in English | MEDLINE | ID: mdl-9278383

ABSTRACT

Activation of transcription factor NF-kappaB involves the signal-dependent degradation of basally phosphorylated inhibitors such as IkappaBalpha and IkappaBbeta. The gene encoding IkappaBalpha is under NF-kappaB control, which provides a negative feedback loop to terminate the induced NF-kappaB response. However, recent studies have identified a hypophosphorylated pool of IkappaBbeta that shields nuclear NF-kappaB from inhibition by newly synthesized IkappaBalpha. In the present work, we provide three lines of evidence indicating that this protection mechanism is regulated by the C-terminal PEST domain of IkappaBbeta. First, disruption of two basal phosphoacceptors present in the IkappaBbeta PEST domain (Ser-313 and Ser-315) yields a mutant that forms ternary complexes with NF-kappaB and its target DNA-binding site. Second, based on in vitro mixing experiments, these ternary complexes are resistant to the inhibitory action of IkappaBalpha. Third, mutants of IkappaBbeta that are defective for phosphorylation at Ser-313 and Ser-315 fail to efficiently block NF-kappaB-directed transcription in vivo, whereas replacement of these two IkappaBbeta residues with a phosphoserine mimetic generates a fully functional repressor. Taken together, our findings suggest that the functional fate of NF-kappaB when bound to IkappaBbeta is critically dependent on the phosphorylation status of the IkappaBbeta PEST domain.


Subject(s)
DNA-Binding Proteins/metabolism , I-kappa B Proteins , NF-kappa B/metabolism , Binding Sites , Cell Line , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Humans , Mutation , NF-kappa B/antagonists & inhibitors , Phosphorylation , Serine/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...