Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 263
Filtrer
1.
Sci Adv ; 7(18)2021 04.
Article de Anglais | MEDLINE | ID: mdl-33931459

RÉSUMÉ

Duchenne muscular dystrophy (DMD) is a fatal muscle disease caused by the lack of dystrophin, which maintains muscle membrane integrity. We used an adenine base editor (ABE) to modify splice donor sites of the dystrophin gene, causing skipping of a common DMD deletion mutation of exon 51 (∆Ex51) in cardiomyocytes derived from human induced pluripotent stem cells, restoring dystrophin expression. Prime editing was also capable of reframing the dystrophin open reading frame in these cardiomyocytes. Intramuscular injection of ∆Ex51 mice with adeno-associated virus serotype-9 encoding ABE components as a split-intein trans-splicing system allowed gene editing and disease correction in vivo. Our findings demonstrate the effectiveness of nucleotide editing for the correction of diverse DMD mutations with minimal modification of the genome, although improved delivery methods will be required before these strategies can be used to sufficiently edit the genome in patients with DMD.


Sujet(s)
Cellules souches pluripotentes induites , Myopathie de Duchenne , Animaux , Systèmes CRISPR-Cas , Dystrophine/génétique , Dystrophine/métabolisme , Exons , Édition de gène , Humains , Cellules souches pluripotentes induites/métabolisme , Souris , Myopathie de Duchenne/génétique , Myopathie de Duchenne/métabolisme , Myopathie de Duchenne/thérapie , Délétion de séquence
2.
Diabetologia ; 55(10): 2682-2692, 2012 Oct.
Article de Anglais | MEDLINE | ID: mdl-22820510

RÉSUMÉ

AIMS/HYPOTHESIS: Activation of the G protein-coupled receptor (GPR)40 by long-chain fatty acids potentiates glucose-stimulated insulin secretion (GSIS) from pancreatic beta cells, and GPR40 agonists are in clinical development for type 2 diabetes therapy. GPR40 couples to the G protein subunit Gα(q/11) but the signalling cascade activated downstream is unknown. This study aimed to determine the mechanisms of GPR40-dependent potentiation of GSIS by fatty acids. METHODS: Insulin secretion in response to glucose, oleate or diacylglycerol (DAG) was assessed in dynamic perifusions and static incubations in islets from wild-type (WT) and Gpr40 (-/-) mice. Depolymerisation of filamentous actin (F-actin) was visualised by phalloidin staining and epifluorescence. Pharmacological and molecular approaches were used to ascertain the roles of protein kinase D (PKD) and protein kinase C delta in GPR40-mediated potentiation of GSIS. RESULTS: Oleate potentiates the second phase of GSIS, and this effect is largely dependent upon GPR40. Accordingly, oleate induces rapid F-actin remodelling in WT but not in Gpr40 (-/-) islets. Exogenous DAG potentiates GSIS in both WT and Gpr40 (-/-) islets. Oleate induces PKD phosphorylation at residues Ser-744/748 and Ser-916 in WT but not Gpr40 (-/-) islets. Importantly, oleate-induced F-actin depolymerisation and potentiation of GSIS are lost upon pharmacological inhibition of PKD1 or deletion of Prkd1. CONCLUSIONS/INTERPRETATION: We conclude that the signalling cascade downstream of GPR40 activation by fatty acids involves activation of PKD1, F-actin depolymerisation and potentiation of second-phase insulin secretion. These results provide important information on the mechanisms of action of GPR40, a novel drug target for type 2 diabetes.


Sujet(s)
Insuline/métabolisme , Ilots pancréatiques/métabolisme , Protéine kinase C/physiologie , Récepteurs couplés aux protéines G/physiologie , Actines/métabolisme , Animaux , Cellules cultivées , Diglycéride/pharmacologie , Glucose/pharmacologie , Sécrétion d'insuline , Ilots pancréatiques/cytologie , Ilots pancréatiques/effets des médicaments et des substances chimiques , Souris , Souris knockout , Modèles animaux , Acide oléique/pharmacologie , Protein kinase C-delta/déficit , Protein kinase C-delta/génétique , Protein kinase C-delta/physiologie , Récepteurs couplés aux protéines G/déficit , Récepteurs couplés aux protéines G/génétique , Transduction du signal/physiologie
3.
J Anim Sci ; 90(1): 171-83, 2012 Jan.
Article de Anglais | MEDLINE | ID: mdl-21856901

RÉSUMÉ

The molecular mechanisms that regulate fat deposition in bovine adipose tissue have not been well studied. To elucidate the genes and gene networks involved in bovine fat development, transcriptional profiles of backfat (BF) tissues from Hereford × Aberdeen Angus (HEAN, n = 6) and Charolais × Red Angus (CHRA, n = 6) steers with high or low BF thickness were characterized by digital gene expression-tag profiling. Approximately 9.8 to 21.9 million tags were obtained for each library, and a total of 18,034 genes were identified. In total, 650 genes were found to be differentially expressed, with a greater than 1.5-fold difference between the 2 crossbreds (Benjamini-Hochberg false discovery rate ≤ 0.05). The majority of differentially expressed genes that were more highly expressed in CHRA vs. HEAN were associated with development, whereas the differentially expressed genes with greater expression in HEAN vs. CHRA were overrepresented in biological processes such as metabolism and immune response. Thirty-six and 152 differentially expressed genes were detected between animals with high (n = 3) and low (n = 3) BF thickness in HEAN and CHRA, respectively (Benjamini-Hochberg false discovery rate ≤0.05). The differentially expressed genes between high and low groups in CHRA were related to cell proliferation and development processes. In addition, lipid metabolism was 1 of the top 5 molecular and cellular functions identified in both crossbreds. Ten and 17 differentially expressed genes were found to be involved in fat metabolism in HEAN and CHRA, respectively. Genes associated with obesity, such as PTX3 (pentraxin 3, long) and SERPINE1 (serpin peptidase inhibitor, clade E, member 1), were more highly expressed (P < 0.05) in the subset of CHRA animals with greater BF thickness. Our study revealed that the expression patterns of genes in BF tissues differed depending on the genetic background of the cattle.


Sujet(s)
Bovins/génétique , Analyse de profil d'expression de gènes , Obésité/médecine vétérinaire , Graisse sous-cutanée/métabolisme , Animaux , Bovins/croissance et développement , Bovins/métabolisme , Croisements génétiques , Réseaux de régulation génique , Mâle , Obésité/génétique , Obésité/métabolisme , Séquençage par oligonucléotides en batterie/médecine vétérinaire , Réaction de polymérisation en chaine en temps réel/médecine vétérinaire , Graisse sous-cutanée/croissance et développement
5.
Annu Rev Physiol ; 65: 45-79, 2003.
Article de Anglais | MEDLINE | ID: mdl-12524460

RÉSUMÉ

Cardiac hypertrophy is the heart's response to a variety of extrinsic and intrinsic stimuli that impose increased biomechanical stress. While hypertrophy can eventually normalize wall tension, it is associated with an unfavorable outcome and threatens affected patients with sudden death or progression to overt heart failure. Accumulating evidence from studies in human patients and animal models suggests that in most instances hypertrophy is not a compensatory response to the change in mechanical load, but rather is a maladaptive process. Accordingly, modulation of myocardial growth without adversely affecting contractile function is increasingly recognized as a potentially auspicious approach in the prevention and treatment of heart failure. In this review, we summarize recent insights into hypertrophic signaling and consider several novel antihypertrophic strategies.


Sujet(s)
Cardiomégalie/métabolisme , Cardiomégalie/physiopathologie , Coeur/physiopathologie , Myocarde/métabolisme , Adaptation physiologique/physiologie , Animaux , Humains
7.
Article de Anglais | MEDLINE | ID: mdl-12858532

RÉSUMÉ

We have established the beginnings of a road map to understand how ventricular cells become specified, differentiate, and expand into a functional cardiac chamber (Fig. 5). The transcriptional networks described here provide clear evidence that disruption of pathways affecting ventricular growth could be the underlying etiology in a subset of children born with malformation of the right or left ventricle. As we learn details of the precise mechanisms through which the critical factors function, the challenge will lie in devising innovative methods to augment or modify the effects of gene mutations on ventricular development. Because most congenital heart disease likely occurs in a setting of heterozygous, predisposing mutations of one or more genes, modulation of activity of critical pathways in a preventive fashion may be useful in averting disease in genetically susceptible individuals.


Sujet(s)
Cardiopathies congénitales/génétique , Hypoplasie du coeur gauche/génétique , Protéines du muscle , Protéines de Xénope , Animaux , Facteurs de transcription à motif basique hélice-boucle-hélice , Plan d'organisation du corps/génétique , Protéines de liaison à l'ADN/déficit , Protéines de liaison à l'ADN/génétique , Régulation de l'expression des gènes au cours du développement , Cardiopathies congénitales/embryologie , Cardiopathies congénitales/physiopathologie , Ventricules cardiaques/malformations , Protéine homéotique Nkx-2.5 , Protéines à homéodomaine/génétique , Protéines à homéodomaine/physiologie , Humains , Hypoplasie du coeur gauche/embryologie , Hypoplasie du coeur gauche/physiopathologie , Souris , Souris knockout , Modèles cardiovasculaires , Facteurs de transcription/déficit , Facteurs de transcription/génétique , Protéines de poisson-zèbre
8.
Dev Cell ; 1(5): 705-15, 2001 Nov.
Article de Anglais | MEDLINE | ID: mdl-11709190

RÉSUMÉ

Somatic muscle formation in Drosophila requires fusion of muscle founder cells with fusion-competent myoblasts. In a genetic screen for genes that control muscle development, we identified antisocial (ants), a gene that encodes an ankyrin repeat-, TPR repeat-, and RING finger-containing protein, required for myoblast fusion. In ants mutant embryos, founder cells and fusion-competent myoblasts are properly specified and patterned, but they are unable to form myotubes. ANTS, which is expressed specifically in founder cells, interacts with the cytoplasmic domain of Dumbfounded, a founder cell transmembrane receptor, and with Myoblast city, a cytoskeletal protein, both of which are also required for myoblast fusion. These findings suggest that ANTS functions as an intracellular adaptor protein that relays signals from Dumbfounded to the cytoskeleton during myoblast fusion.


Sujet(s)
Protéines du cytosquelette , Drosophila/embryologie , Protéines membranaires , Protéines du muscle/composition chimique , Protéines du muscle/métabolisme , Muscles/cytologie , Muscles/embryologie , Motifs d'acides aminés , Séquence d'acides aminés , Animaux , Différenciation cellulaire , Fusion cellulaire , Taille de la cellule , Cellules cultivées , Clonage moléculaire , Cytoplasme/composition chimique , Cytoplasme/métabolisme , Drosophila/cytologie , Drosophila/génétique , Protéines de Drosophila/composition chimique , Protéines de Drosophila/génétique , Protéines de Drosophila/métabolisme , Embryon de mammifère/cytologie , Embryon de mammifère/métabolisme , Embryon non mammalien , Régulation de l'expression des gènes au cours du développement , Protéines d'insecte/composition chimique , Protéines d'insecte/génétique , Protéines d'insecte/métabolisme , Mésoderme/métabolisme , Souris , Données de séquences moléculaires , Protéines du muscle/génétique , Mutation , Liaison aux protéines , ARN messager/génétique , ARN messager/métabolisme , Séquences répétées d'acides aminés , Cellules souches/cytologie , Cellules souches/métabolisme
9.
EMBO J ; 20(22): 6414-23, 2001 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-11707412

RÉSUMÉ

Gene expression in skeletal muscles of adult vertebrates is altered profoundly by changing patterns of contractile work. Here we observed that the functional activity of MEF2 transcription factors is stimulated by sustained periods of endurance exercise or motor nerve pacing, as assessed by expression in trans genic mice of a MEF2-dependent reporter gene (desMEF2-lacZ). This response is accompanied by transformation of specialized myofiber subtypes, and is blocked either by cyclosporin A, a specific chemical inhibitor of calcineurin, or by forced expression of the endogenous calcineurin inhibitory protein, myocyte-enriched calcineurin interacting protein 1. Calcineurin removes phosphate groups from MEF2, and augments the potency of the transcriptional activation domain of MEF2 fused to a heterologous DNA binding domain. Across a broad range, the enzymatic activity of calcineurin correlates directly with expression of endogenous genes that are transcriptionally activated by muscle contractions. These results delineate a molecular pathway in which calcineurin and MEF2 participate in the adaptive mechanisms by which skeletal myofibers acquire specialized contractile and metabolic properties as a function of changing patterns of muscle contraction.


Sujet(s)
Calcineurine/métabolisme , Protéines de liaison à l'ADN/métabolisme , Facteurs de transcription/métabolisme , Animaux , Ciclosporine/pharmacologie , ADN complémentaire/métabolisme , Relation dose-effet des médicaments , Activation enzymatique , Antienzymes/pharmacologie , Gènes rapporteurs , Immunotransfert , Cinétique , Facteurs de transcription MEF2 , Souris , Souris de lignée C57BL , Souris transgéniques , Modèles biologiques , Contraction musculaire , Muscles squelettiques/métabolisme , Facteurs de régulation myogènes , Myoglobine/biosynthèse , Conditionnement physique d'animal , Effort physique , Plasmides/métabolisme , Tests aux précipitines , Liaison aux protéines , Structure tertiaire des protéines , ARN/métabolisme , ARN messager/métabolisme , Facteurs temps , Transcription génétique , Activation de la transcription , Transfection , beta-Galactosidase/métabolisme
10.
Genes Dev ; 15(22): 3039-49, 2001 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-11711438

RÉSUMÉ

Neural crest cells play a key role in craniofacial development. The endothelin family of secreted polypeptides regulates development of several neural crest sublineages, including the branchial arch neural crest. The basic helix-loop-helix transcription factor dHAND is also required for craniofacial development, and in endothelin-1 (ET-1) mutant embryos, dHAND expression in the branchial arches is down-regulated, implicating it as a transcriptional effector of ET-1 action. To determine the mechanism that links ET-1 signaling to dHAND transcription, we analyzed the dHAND gene for cis-regulatory elements that control transcription in the branchial arches. We describe an evolutionarily conserved dHAND enhancer that requires ET-1 signaling for activity. This enhancer contains four homeodomain binding sites that are required for branchial arch expression. By comparing protein binding to these sites in branchial arch extracts from endothelin receptor A (EdnrA) mutant and wild-type mouse embryos, we identified Dlx6, a member of the Distal-less family of homeodomain proteins, as an ET-1-dependent binding factor. Consistent with this conclusion, Dlx6 was down-regulated in branchial arches from EdnrA mutant mice. These results suggest that Dlx6 acts as an intermediary between ET-1 signaling and dHAND transcription during craniofacial morphogenesis.


Sujet(s)
Protéines de liaison à l'ADN/métabolisme , Endothéline-1/métabolisme , Protéines à homéodomaine/métabolisme , Protéines à homéodomaine/physiologie , Facteurs de transcription/métabolisme , Animaux , Séquence nucléotidique , Facteurs de transcription à motif basique hélice-boucle-hélice , Sites de fixation , Cellules COS , Embryon de poulet , Protéines de liaison à l'ADN/composition chimique , Régulation négative , Délétion de gène , Régulation de l'expression des gènes , Gènes rapporteurs , Hybridation in situ , Souris , Souris transgéniques , Modèles génétiques , Données de séquences moléculaires , Liaison aux protéines , Structure tertiaire des protéines , Transduction du signal , Facteurs de transcription/composition chimique , Transcription génétique , Protéines de poisson-zèbre
11.
Development ; 128(22): 4623-33, 2001 Nov.
Article de Anglais | MEDLINE | ID: mdl-11714687

RÉSUMÉ

Members of the MEF2 family of transcription factors are upregulated during skeletal muscle differentiation and cooperate with the MyoD family of myogenic basic helix-loop-helix (bHLH) transcription factors to control the expression of muscle-specific genes. To determine the mechanisms that regulate MEF2 gene expression during skeletal muscle development, we analyzed the mouse Mef2c gene for cis-regulatory elements that direct expression in the skeletal muscle lineage in vivo. We describe a skeletal muscle-specific control region for Mef2c that is sufficient to direct lacZ reporter gene expression in a pattern that recapitulates that of the endogenous Mef2c gene in skeletal muscle during pre- and postnatal development. This control region is a direct target for the binding of myogenic bHLH and MEF2 proteins. Mutagenesis of the Mef2c control region shows that a binding site for myogenic bHLH proteins is essential for expression at all stages of skeletal muscle development, whereas an adjacent MEF2 binding site is required for maintenance but not for initiation of Mef2c transcription. Our findings reveal the existence of a regulatory circuit between these two classes of transcription factors that induces, amplifies and maintains their expression during skeletal muscle development.


Sujet(s)
Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Développement musculaire/génétique , Muscles squelettiques/embryologie , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Animaux , Séquence nucléotidique , Éléments activateurs (génétique) , Régulation de l'expression des gènes au cours du développement , Facteurs de transcription MEF2 , Souris , Souris transgéniques , Données de séquences moléculaires , Protéine MyoD/métabolisme , Facteurs de régulation myogènes/génétique , Facteurs de régulation myogènes/métabolisme , Séquences d'acides nucléiques régulatrices
12.
Circ Res ; 89(11): 997-1004, 2001 Nov 23.
Article de Anglais | MEDLINE | ID: mdl-11717156

RÉSUMÉ

beta-Adrenergic receptor (betaAR) signaling, which elevates intracellular cAMP and enhances cardiac contractility, is severely impaired in the failing heart. Protein kinase A (PKA) is activated by cAMP, but the long-term physiological effect of PKA activation on cardiac function is unclear. To investigate the consequences of chronic cardiac PKA activation in the absence of upstream events associated with betaAR signaling, we generated transgenic mice that expressed the catalytic subunit of PKA in the heart. These mice developed dilated cardiomyopathy with reduced cardiac contractility, arrhythmias, and susceptibility to sudden death. As seen in human heart failure, these abnormalities correlated with PKA-mediated hyperphosphorylation of the cardiac ryanodine receptor/Ca(2+)-release channel, which enhances Ca(2+) release from the sarcoplasmic reticulum, and phospholamban, which regulates the sarcoplasmic reticulum Ca(2+)-ATPase. These findings demonstrate a specific role for PKA in the pathogenesis of heart failure, independent of more proximal events in betaAR signaling, and support the notion that PKA activity is involved in the adverse effects of chronic betaAR signaling.


Sujet(s)
Cardiomyopathie dilatée/étiologie , Cyclic AMP-Dependent Protein Kinases/métabolisme , Cyclic AMP-Dependent Protein Kinases/physiologie , Mort subite cardiaque/étiologie , Animaux , Protéines de liaison au calcium/métabolisme , Cardiomyopathie dilatée/enzymologie , Cardiomyopathie dilatée/métabolisme , Cyclic AMP-Dependent Protein Kinases/génétique , Activation enzymatique , Humains , Souris , Souris transgéniques , Contraction myocardique , Chaînes lourdes de myosine/génétique , Phosphorylation , Canal de libération du calcium du récepteur à la ryanodine/métabolisme
13.
Development ; 128(19): 3855-66, 2001 Oct.
Article de Anglais | MEDLINE | ID: mdl-11585810

RÉSUMÉ

Little is known about the genesis and patterning of tendons and other connective tissues, mostly owing to the absence of early markers. We have found that Scleraxis, a bHLH transcription factor, is a highly specific marker for all the connective tissues that mediate attachment of muscle to bone in chick and mouse, including the limb tendons, and show that early scleraxis expression marks the progenitor cell populations for these tissues. In the early limb bud, the tendon progenitor population is found in the superficial proximomedial mesenchyme. Using the scleraxis gene as a marker we show that these progenitors are induced by ectodermal signals and restricted by bone morphogenetic protein (BMP) signaling within the mesenchyme. Application of Noggin protein antagonizes this endogenous BMP activity and induces ectopic scleraxis expression. However, the presence of excess tendon progenitors does not lead to the production of additional or longer tendons, indicating that additional signals are required for the final formation of a tendon. Finally, we show that the endogenous expression of noggin within the condensing digit cartilage contributes to the induction of distal tendons.


Sujet(s)
Tendons/cytologie , Tendons/embryologie , Facteurs de transcription/métabolisme , Animaux , Protéines aviaires , Facteurs de transcription à motif basique hélice-boucle-hélice , Marqueurs biologiques , Protéines morphogénétiques osseuses/métabolisme , Protéines de transport , Embryon de poulet , Tissu conjonctif/embryologie , Tissu conjonctif/métabolisme , Ectoderme/métabolisme , Régulation de l'expression des gènes au cours du développement , Bourgeons de membre/cytologie , Bourgeons de membre/métabolisme , Protéines/métabolisme , Transduction du signal , Cellules souches/métabolisme , Tendons/métabolisme , Facteurs de transcription/génétique
14.
Curr Opin Genet Dev ; 11(5): 497-504, 2001 Oct.
Article de Anglais | MEDLINE | ID: mdl-11532390

RÉSUMÉ

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.


Sujet(s)
Acetyltransferases/génétique , Acetyltransferases/métabolisme , Histone deacetylases/métabolisme , Muscles squelettiques/enzymologie , Muscles squelettiques/croissance et développement , Protéines de Saccharomyces cerevisiae , Transport nucléaire actif , Animaux , Protéines de transport , Protéines du cycle cellulaire/métabolisme , Chromatine/enzymologie , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Histone acetyltransferases , Humains , Facteurs de transcription MEF2 , Développement musculaire/génétique , Muscles squelettiques/cytologie , Muscles squelettiques/métabolisme , Protéine MyoD/génétique , Protéine MyoD/métabolisme , Facteurs de régulation myogènes , Corépresseur-2 de récepteur nucléaire , Coactivateur-2 de récepteur nucléaire , Phosphoprotéines/métabolisme , Protéines de répression/métabolisme , Transduction du signal , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Facteurs de transcription CBP-p300
15.
Mol Cell Biol ; 21(18): 6312-21, 2001 Sep.
Article de Anglais | MEDLINE | ID: mdl-11509672

RÉSUMÉ

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.


Sujet(s)
Histone deacetylases/génétique , Protéines de répression , Séquence d'acides aminés , Animaux , Cellules COS , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Histone Deacetylase 2 , Histone deacetylases/métabolisme , Facteurs de transcription MEF2 , Données de séquences moléculaires , Facteurs de régulation myogènes , Délétion de séquence , Transduction du signal/génétique , Relation structure-activité , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme
16.
Cell ; 105(7): 851-62, 2001 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-11439182

RÉSUMÉ

Serum response factor (SRF) regulates transcription of numerous muscle and growth factor-inducible genes. Because SRF is not muscle specific, it has been postulated to activate muscle genes by recruiting myogenic accessory factors. Using a bioinformatics-based screen for unknown cardiac-specific genes, we identified a novel and highly potent transcription factor, named myocardin, that is expressed in cardiac and smooth muscle cells. Myocardin belongs to the SAP domain family of nuclear proteins and activates cardiac muscle promoters by associating with SRF. Expression of a dominant negative mutant of myocardin in Xenopus embryos interferes with myocardial cell differentiation. Myocardin is the founding member of a class of muscle transcription factors and provides a mechanism whereby SRF can convey myogenic activity to cardiac muscle genes.


Sujet(s)
Biologie informatique , Protéines de liaison à l'ADN/métabolisme , Myocarde/métabolisme , Protéines nucléaires/métabolisme , Transactivateurs/métabolisme , Activation de la transcription/génétique , Séquence d'acides aminés , Animaux , Cellules COS , Protéines de liaison à l'ADN/génétique , Embryon de mammifère/physiologie , Embryon non mammalien , Étiquettes de séquences exprimées , Gènes rapporteurs/génétique , Souris , Microinjections , Données de séquences moléculaires , Muscles lisses/physiologie , Protéines nucléaires/composition chimique , Protéines nucléaires/génétique , Régions promotrices (génétique)/génétique , Liaison aux protéines , Structure tertiaire des protéines , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/métabolisme , Alignement de séquences , Facteur de réponse au sérum , Transactivateurs/composition chimique , Transactivateurs/génétique , Xenopus laevis/embryologie , Xenopus laevis/génétique
17.
Dev Biol ; 234(2): 497-509, 2001 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-11397016

RÉSUMÉ

MEF2C is a MADS-box transcription factor required for cardiac myogenesis and morphogenesis. In MEF2C mutant mouse embryos, heart development arrests at the looping stage (embryonic day 9.0), the future right ventricular chamber fails to form, and cardiomyocyte differentiation is disrupted. To identify genes regulated by MEF2C in the developing heart, we performed differential array analysis coupled with subtractive cloning using RNA from heart tubes of wild-type and MEF2C-null embryos. Here, we describe a novel MEF2C-dependent gene that encodes a cardiac-restricted protein, called CHAMP (cardiac helicase activated by MEF2 protein), that contains seven conserved motifs characteristic of helicases involved in RNA processing, DNA replication, and transcription. During mouse embryogenesis, CHAMP expression commences in the linear heart tube at embryonic day 8.0, shortly after initiation of MEF2C expression in the cardiogenic region. Thereafter, CHAMP is expressed specifically in embryonic and postnatal cardiomyocytes. At the trabeculation stage of heart development, CHAMP expression is highest in the trabecular region in which cardiomyocytes have exited the cell cycle and is lowest in the proliferative compact zone. These findings suggest that CHAMP acts downstream of MEF2C in a cardiac-specific regulatory pathway for RNA processing and/or transcriptional control.


Sujet(s)
Helicase/génétique , Coeur/embryologie , Facteurs de régulation myogènes/métabolisme , RNA helicases , Séquence d'acides aminés , Animaux , Cardiomégalie/étiologie , Compartimentation cellulaire , Clonage moléculaire/méthodes , Régulation négative , Régulation de l'expression des gènes au cours du développement , Facteurs de transcription MEF2 , Souris , Souches mutantes de souris , Données de séquences moléculaires , Spécificité d'organe , Similitude de séquences d'acides aminés , Distribution tissulaire
18.
EMBO J ; 20(11): 2757-67, 2001 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-11387209

RÉSUMÉ

Mitogen-activated protein kinase (MAPK) pathways couple intrinsic and extrinsic signals to hypertrophic growth of cardiomyocytes. The MAPK kinase MEK5 activates the MAPK ERK5. To investigate the potential involvement of MEK5-ERK5 in cardiac hypertrophy, we expressed constitutively active and dominant-negative forms of MEK5 in cardiomyocytes in vitro. MEK5 induced a form of hypertrophy in which cardiomyocytes acquired an elongated morphology and sarcomeres were assembled in a serial manner. The cytokine leukemia inhibitory factor (LIF), which stimulates MEK5 activity, evoked a similar response. Moreover, a dominant-negative MEK5 mutant specifically blocked LIF-induced elongation of cardiomyocytes and reduced expression of fetal cardiac genes without blocking other aspects of LIF-induced hypertrophy. Consistent with the ability of MEK5 to induce serial assembly of sarcomeres in vitro, cardiac-specific expression of activated MEK5 in transgenic mice resulted in eccentric cardiac hypertrophy that progressed to dilated cardiomyopathy and sudden death. These findings reveal a specific role for MEK5-ERK5 in the induction of eccentric cardiac hypertrophy and in transduction of cytokine signals that regulate serial sarcomere assembly.


Sujet(s)
Cardiomyopathie dilatée/physiopathologie , Coeur/physiopathologie , Interleukine-6 , Mitogen-Activated Protein Kinase Kinases/métabolisme , Mitogen-Activated Protein Kinases/métabolisme , Myocarde/enzymologie , Sarcomères/physiologie , Animaux , Animaux nouveau-nés , Apoptose , Cellules COS , Cardiomyopathie dilatée/génétique , Cardiomyopathie dilatée/anatomopathologie , Cellules cultivées , Chlorocebus aethiops , Activation enzymatique , Inhibiteurs de croissance/pharmacologie , Ventricules cardiaques , Humains , Facteur inhibiteur de la leucémie , Lymphokines/pharmacologie , MAP Kinase Kinase 5 , Souris , Souris transgéniques , Mitogen-Activated Protein Kinase 7 , Mitogen-Activated Protein Kinase Kinases/génétique , Myocarde/cytologie , Myocarde/anatomopathologie , Rats , Protéines recombinantes/métabolisme , Délétion de séquence , Transfection
19.
Proc Natl Acad Sci U S A ; 98(13): 7354-9, 2001 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-11390982

RÉSUMÉ

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.


Sujet(s)
Protéines de transport/génétique , Protéines de transport/métabolisme , Développement embryonnaire et foetal , Régulation de l'expression des gènes au cours du développement , Muscles squelettiques/physiologie , Protéines 14-3-3 , Animaux , Cellules COS , Différenciation cellulaire , Lignée cellulaire , Chlorocebus aethiops , Protéines de liaison à l'ADN/métabolisme , Histone deacetylases/métabolisme , Facteurs de transcription MEF2 , Souris , Muscles squelettiques/cytologie , Muscles squelettiques/embryologie , Protéine MyoD/métabolisme , Facteurs de régulation myogènes , Protéines recombinantes/métabolisme , Protéines de répression/génétique , Protéines de répression/métabolisme , Transduction du signal , Facteurs de transcription/métabolisme , Transcription génétique , Transfection , Tyrosine 3-monooxygenase/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE