Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Hum Gene Ther ; 35(15-16): 543-554, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38970421

RESUMEN

Adeno-associated virus-mediated gene therapies for certain muscle disorders require regulatory cassettes that provide high-level, striated muscle-specific activity. However, cardiotoxicity has emerged as a serious concern in clinical trials for Duchenne muscular dystrophy and X-linked myotubular myopathy. While this may be caused by systemic inflammatory effects of the treatment, high transgene expression in the heart may also play a role. Thus, certain muscle disorders may require a modulated level of therapeutic expression in the heart, while others may not require any cardiac expression at all. Additionally, the size of some cargos requires regulatory cassettes to be small enough that large cDNAs and other therapeutic payloads can be accommodated. Thus, we have performed enhancer/promoter optimization to develop highly minimized regulatory cassettes that are active in skeletal muscles, with either low or no detectable activity in cardiac muscle. Our No-heart (NH) cassette is active in most skeletal muscles, but exhibits only very low activity in extensor digitorum longus (EDL), soleus, and diaphragm, and no activity in the heart. By contrast, our Have a Little Heart (HLH) cassette displays high activity in most skeletal muscles, comparable to the ∼800-bp CK8 cassette, with increased activity in EDL, soleus, and diaphragm, and low activity in the heart. Due to their small size, these cassettes can be used in therapeutic strategies with both flexible (e.g., antisense) and stringent (e.g., CRISPR/Cas or bicistronic) size limitations. Thus, our new cassettes may be useful for gene therapies of muscle disorders in which the need for low or almost no expression in cardiac muscle would outweigh the need for high levels of therapeutic product in certain skeletal muscles.


Asunto(s)
Dependovirus , Terapia Genética , Vectores Genéticos , Músculo Esquelético , Miocardio , Músculo Esquelético/metabolismo , Animales , Dependovirus/genética , Miocardio/metabolismo , Terapia Genética/métodos , Humanos , Vectores Genéticos/genética , Ratones , Regiones Promotoras Genéticas , Transgenes , Elementos de Facilitación Genéticos
2.
J Neuromuscul Dis ; 9(1): 83-93, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34459413

RESUMEN

BACKGROUND: Facioscapulohumeral muscular dystrophy (FSHD) is one of the most common myopathies in adults, displaying a progressive, frequently asymmetric involvement of a typical muscles' pattern. FSHD is associated with epigenetic derepression of the polymorphic D4Z4 repeat on chromosome 4q, leading to DUX4 retrogene toxic expression in skeletal muscles. Identifying biomarkers that correlate with disease severity would facilitate clinical management and assess potential FSHD therapeutics' efficacy. OBJECTIVES: This study purpose was to analyze serum cytokines to identify potential biomarkers in a large cohort of adult patients with FSHD. METHODS: We retrospectively measured the levels of 20 pro-inflammatory and regulatory cytokines in sera from 100 genetically confirmed adult FSHD1 patients. Associations between cytokine concentrations and various clinical scores were investigated. We then measured serum and muscle interleukin 6 (IL-6) levels in a validated FSHD-like mouse model, ranging in severity and DUX4 expression. RESULTS: IL-6 was identified as the only cytokine with a concentration correlating with several clinical severity and functional scores, including Clinical Severity Score, Manual Muscle Testing sum score, Brooke and Vignos scores. Further, FSHD patients displayed overall IL-6 levels more than twice high as control, and patients with milder phenotypes exhibited lower IL-6 serum concentration than those with severe muscular weakness. Lastly, an FSHD-like mouse model analysis confirmed that IL-6 levels positively correlate with disease severity and DUX4 expression. CONCLUSIONS: Serum IL-6, therefore, shows promise as a serum biomarker of FSHD severity in a large cohort of FSHD1 adult patients.


Asunto(s)
Interleucina-6/sangre , Distrofia Muscular Facioescapulohumeral/sangre , Distrofia Muscular Facioescapulohumeral/diagnóstico , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Biomarcadores , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Estudios Retrospectivos , Índice de Severidad de la Enfermedad , Adulto Joven
3.
Mol Ther Methods Clin Dev ; 20: 298-311, 2021 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-33511244

RESUMEN

Facioscapulohumeral muscular dystrophy (FSHD) is caused by incomplete silencing of the disease locus, leading to pathogenic misexpression of DUX4 in skeletal muscle. Previously, we showed that CRISPR inhibition could successfully target and repress DUX4 in FSHD myocytes. However, an effective therapy will require both efficient delivery of therapeutic components to skeletal muscles and long-term repression of the disease locus. Thus, we re-engineered our platform to allow in vivo delivery of more potent epigenetic repressors. We designed an FSHD-optimized regulatory cassette to drive skeletal muscle-specific expression of dCas9 from Staphylococcus aureus fused to HP1α, HP1γ, the MeCP2 transcriptional repression domain, or the SUV39H1 SET domain. Targeting each regulator to the DUX4 promoter/exon 1 increased chromatin repression at the locus, specifically suppressing DUX4 and its target genes in FSHD myocytes and in a mouse model of the disease. Importantly, minimizing the regulatory cassette and using the smaller Cas9 ortholog allowed our therapeutic cassettes to be effectively packaged into adeno-associated virus (AAV) vectors for in vivo delivery. By engineering a muscle-specific epigenetic CRISPR platform compatible with AAV vectors for gene therapy, we have laid the groundwork for clinical use of dCas9-based chromatin effectors in skeletal muscle disorders.

4.
Dev Cell ; 50(5): 525-526, 2019 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-31505171

RESUMEN

In this issue of Developmental Cell, Chew et al. (2019) show that the pioneer factor DUX4 is misexpressed in tumors, where it suppresses anti-tumor immune activity. Their findings provide a new mechanism for immune evasion in cancer and highlight the pathogenic effects of re-expressing an embryonic program in adult cells.


Asunto(s)
Distrofia Muscular Facioescapulohumeral , Neoplasias , Proteínas de Homeodominio , Humanos , Evasión Inmune
5.
Mol Ther ; 26(7): 1797-1807, 2018 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-29759937

RESUMEN

Facioscapulohumeral muscular dystrophy (FSHD) is caused by epigenetic de-repression of the disease locus, leading to pathogenic misexpression of the DUX4 gene in skeletal muscle. While the factors and pathways involved in normal repression of the FSHD locus in healthy cells have been well characterized, very little is known about those responsible for the aberrant activation of DUX4-fl in FSHD myocytes. Reasoning that DUX4-fl activators might represent useful targets for small molecule inhibition, we performed a highly targeted, candidate-based screen of epigenetic regulators in primary FSHD myocytes. We confirmed several of the strongest and most specific candidates (ASH1L, BRD2, KDM4C, and SMARCA5) in skeletal myocytes from two other unrelated FSHD1 patients, and we showed that knockdown led to reduced levels of DUX4-fl and DUX4-FL target genes, as well as altered chromatin at the D4Z4 locus. As a second mode of validation, targeting the CRISPR/dCas9-KRAB transcriptional repressor to the promoters of several candidates also led to reduced levels of DUX4-fl. Furthermore, these candidates can be repressed by different methods in skeletal myocytes without major effects on certain critical muscle genes. Our results demonstrate that expression of DUX4-fl is regulated by multiple epigenetic pathways, and they indicate viable, druggable candidates for therapeutic target development.


Asunto(s)
Epigénesis Genética/genética , Proteínas de Homeodominio/genética , Distrofia Muscular Facioescapulohumeral/genética , Distrofia Muscular Facioescapulohumeral/terapia , Adenosina Trifosfatasas/genética , Línea Celular , Cromatina/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Células HEK293 , Humanos , Células Musculares/patología , Fibras Musculares Esqueléticas/fisiología , Músculo Esquelético/fisiología , Regiones Promotoras Genéticas/genética , Transcripción Genética/genética
6.
Antimicrob Agents Chemother ; 59(3): 1534-41, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25534740

RESUMEN

Previous studies indicated that inhibition of efflux pumps augments tuberculosis therapy. In this study, we used timcodar (formerly VX-853) to determine if this efflux pump inhibitor could increase the potency of antituberculosis (anti-TB) drugs against Mycobacterium tuberculosis in in vitro and in vivo combination studies. When used alone, timcodar weakly inhibited M. tuberculosis growth in broth culture (MIC, 19 µg/ml); however, it demonstrated synergism in drug combination studies with rifampin, bedaquiline, and clofazimine but not with other anti-TB agents. When M. tuberculosis was cultured in host macrophage cells, timcodar had about a 10-fold increase (50% inhibitory concentration, 1.9 µg/ml) in the growth inhibition of M. tuberculosis and demonstrated synergy with rifampin, moxifloxacin, and bedaquiline. In a mouse model of tuberculosis lung infection, timcodar potentiated the efficacies of rifampin and isoniazid, conferring 1.0 and 0.4 log10 reductions in bacterial burden in lung, respectively, compared to the efficacy of each drug alone. Furthermore, timcodar reduced the likelihood of a relapse infection when evaluated in a mouse model of long-term, chronic infection with treatment with a combination of rifampin, isoniazid, and timcodar. Although timcodar had no effect on the pharmacokinetics of rifampin in plasma and lung, it did increase the plasma exposure of bedaquiline. These data suggest that the antimycobacterial drug-potentiating activity of timcodar is complex and drug dependent and involves both bacterial and host-targeted mechanisms. Further study of the improvement of the potency of antimycobacterial drugs and drug candidates when used in combination with timcodar is warranted.


Asunto(s)
Antituberculosos/farmacología , Piridinas/farmacología , Animales , Antituberculosos/farmacocinética , Línea Celular , Sinergismo Farmacológico , Femenino , Humanos , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Mycobacterium tuberculosis/efectos de los fármacos
7.
Neuromuscul Disord ; 23(12): 975-80, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24128691

RESUMEN

FSHD2 is a rare form of facioscapulohumeral muscular dystrophy (FSHD) characterized by the absence of a contraction in the D4Z4 macrosatellite repeat region on chromosome 4q35 that is the hallmark of FSHD1. However, hypomethylation of this region is common to both subtypes. Recently, mutations in SMCHD1 combined with a permissive 4q35 allele were reported to cause FSHD2. We identified a novel p.Lys275del SMCHD1 mutation in a family affected with FSHD2 using whole-exome sequencing and linkage analysis. This mutation alters a highly conserved amino acid in the ATPase domain of SMCHD1. Subject III-11 is a male who developed asymmetrical muscle weakness characteristic of FSHD at 13 years. Physical examination revealed marked bilateral atrophy at biceps brachii, bilateral scapular winging, some asymmetrical weakness at tibialis anterior and peroneal muscles, and mild lower facial weakness. Biopsy of biceps brachii in subject II-5, the father of III-11, demonstrated lobulated fibers and dystrophic changes. Endomysial and perivascular inflammation was found, which has been reported in FSHD1 but not FSHD2. Given the previous report of SMCHD1 mutations in FSHD2 and the clinical presentations consistent with the FSHD phenotype, we conclude that the SMCHD1 mutation is the likely cause of the disease in this family.


Asunto(s)
Proteínas Cromosómicas no Histona/genética , Exoma/genética , Distrofia Muscular Facioescapulohumeral/genética , Adolescente , Citocinas/metabolismo , Análisis Mutacional de ADN , Salud de la Familia , Humanos , Masculino , Fenotipo
8.
Circ Res ; 112(3): 432-40, 2013 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-23250985

RESUMEN

RATIONALE: Human genetics have implicated the 5-lipoxygenase enzyme in the pathogenesis of cardiovascular disease, and an inhibitor of the 5-lipoxygenase activating protein (FLAP) is in clinical development for asthma. OBJECTIVE: Here we determined whether FLAP deletion modifies the response to vascular injury. METHODS AND RESULTS: Vascular remodeling was characterized 4 weeks after femoral arterial injury in FLAP knockout mice and wild-type controls. Both neointimal hyperplasia and the intima/media ratio of the injured artery were significantly reduced in the FLAP knockouts, whereas endothelial integrity was preserved. Lesional myeloid cells were depleted and vascular smooth muscle cell (VSMC) proliferation, as reflected by bromodeoxyuridine incorporation, was markedly attenuated by FLAP deletion. Inflammatory cytokine release from FLAP knockout macrophages was depressed, and their restricted ability to induce VSMC migration ex vivo was rescued with leukotriene B(4). FLAP deletion restrained injury and attenuated upregulation of the extracellular matrix protein, tenascin C, which affords a scaffold for VSMC migration. Correspondingly, the phenotypic modulation of VSMC to a more synthetic phenotype, reflected by morphological change, loss of α-smooth muscle cell actin, and upregulation of vascular cell adhesion molecule-1 was also suppressed in FLAP knockout mice. Transplantation of FLAP-replete myeloid cells rescued the proliferative response to vascular injury. CONCLUSIONS: Expression of lesional FLAP in myeloid cells promotes leukotriene B(4)-dependent VSMC phenotypic modulation, intimal migration, and proliferation.


Asunto(s)
Proteínas Activadoras de la 5-Lipooxigenasa/metabolismo , Movimiento Celular , Proliferación Celular , Músculo Liso Vascular/enzimología , Células Mieloides/enzimología , Miocitos del Músculo Liso/enzimología , Lesiones del Sistema Vascular/prevención & control , Proteínas Activadoras de la 5-Lipooxigenasa/deficiencia , Proteínas Activadoras de la 5-Lipooxigenasa/genética , Animales , Trasplante de Médula Ósea , Células Cultivadas , Cisteína/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Arteria Femoral/enzimología , Arteria Femoral/lesiones , Arteria Femoral/patología , Genotipo , Hiperplasia , Mediadores de Inflamación/metabolismo , Leucotrieno B4/metabolismo , Leucotrienos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/inmunología , Músculo Liso Vascular/lesiones , Músculo Liso Vascular/patología , Células Mieloides/inmunología , Células Mieloides/trasplante , Miocitos del Músculo Liso/inmunología , Miocitos del Músculo Liso/patología , Neointima , Fenotipo , Tenascina/metabolismo , Factores de Tiempo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Lesiones del Sistema Vascular/enzimología , Lesiones del Sistema Vascular/genética , Lesiones del Sistema Vascular/inmunología , Lesiones del Sistema Vascular/patología
9.
Int J Clin Exp Pathol ; 3(4): 386-400, 2010 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-20490329

RESUMEN

The genetic lesion leading to facioscapulohumeral muscular dystrophy (FSHD) is a dominant deletion at the 4q35 locus. The generally accepted disease model involves an epigenetic dysregulation in the region resulting in the upregulation of one or more proximal genes whose overexpression specifically affects skeletal muscle. However, multiple FSHD candidate genes have been proposed without clear consensus. Using Xenopus laevis as a model for vertebrate development our lab has studied the effects of overexpression of the FSHD candidate gene ortholog, frg1 (FSHD region gene 1), showing that increased levels of frg1 systemically led specifically to an abnormal musculature and increased angiogenesis, the two most prominent clinical features of FSHD. Here we studied the overexpression effects of three other promising FSHD candidate genes, DUX4, DUX4c, and PITX1 using the same model system and methods for direct comparison. Expression of even very low levels of either DUX4 or pitx1 early in development led to massive cellular loss and severely abnormal development. These abnormalities were not muscle specific. In contrast, elevated levels of DUX4c resulted in no detectable adverse affects on muscle and DUX4c levels did not alter the expression of myogenic regulators. This data supports a model for DUX4 and PITX1 in FSHD only as pro-apoptotic factors if their expression in FSHD is confined to cells within the myogenic pathway; neither could account for the vascular pathology prevalent in FSHD. Taken together, increased frg1 expression alone leads to a phenotype that most closely resembles the pathophysiology observed in FSHD patients.


Asunto(s)
Proteínas de Homeodominio/genética , Desarrollo de Músculos/genética , Músculo Esquelético/embriología , Distrofia Muscular Facioescapulohumeral/genética , Factores de Transcripción Paired Box/genética , Animales , Apoptosis/fisiología , Diferenciación Celular , Expresión Génica , Perfilación de la Expresión Génica , Proteínas de Homeodominio/biosíntesis , Humanos , Inmunohistoquímica , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Músculo Esquelético/citología , Músculos , Distrofia Muscular Facioescapulohumeral/metabolismo , Factores de Transcripción Paired Box/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis
10.
J Exp Med ; 205(10): 2409-17, 2008 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-18809715

RESUMEN

The current goal of diabetes therapy is to reduce time-averaged mean levels of glycemia, measured as HbA1c, to prevent diabetic complications. However, HbA1c only explains <25% of the variation in risk of developing complications. Because HbA1c does not correlate with glycemic variability when adjusted for mean blood glucose, we hypothesized that transient spikes of hyperglycemia may be an HbA1c-independent risk factor for diabetic complications. We show that transient hyperglycemia induces long-lasting activating epigenetic changes in the promoter of the nuclear factor kappaB (NF-kappaB) subunit p65 in aortic endothelial cells both in vitro and in nondiabetic mice, which cause increased p65 gene expression. Both the epigenetic changes and the gene expression changes persist for at least 6 d of subsequent normal glycemia, as do NF-kappaB-induced increases in monocyte chemoattractant protein 1 and vascular cell adhesion molecule 1 expression. Hyperglycemia-induced epigenetic changes and increased p65 expression are prevented by reducing mitochondrial superoxide production or superoxide-induced alpha-oxoaldehydes. These results highlight the dramatic and long-lasting effects that short-term hyperglycemic spikes can have on vascular cells and suggest that transient spikes of hyperglycemia may be an HbA1c-independent risk factor for diabetic complications.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus/metabolismo , Epigénesis Genética , Regulación de la Expresión Génica , Glucosa/metabolismo , Hiperglucemia/metabolismo , Animales , Bovinos , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Complicaciones de la Diabetes , Células Endoteliales/citología , Células Endoteliales/fisiología , Hemoglobina Glucada/genética , Hemoglobina Glucada/metabolismo , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Canales Iónicos/genética , Canales Iónicos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Regiones Promotoras Genéticas , Proteína Metiltransferasas , Especies Reactivas de Oxígeno/metabolismo , Factores de Riesgo , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Proteína Desacopladora 1 , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
11.
Science ; 314(5799): 645-7, 2006 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-17068262

RESUMEN

DNA methylation systems are well characterized in vertebrates, but methylation in Drosophila melanogaster and other invertebrates remains controversial. Using the recently sequenced honey bee genome, we present a bioinformatic, molecular, and biochemical characterization of a functional DNA methylation system in an insect. We report on catalytically active orthologs of the vertebrate DNA methyltransferases Dnmt1 and Dnmt3a and b, two isoforms that contain a methyl-DNA binding domain, genomic 5-methyl-deoxycytosine, and CpG-methylated genes. The honey bee provides an opportunity to study the roles of methylation in social contexts.


Asunto(s)
Abejas/genética , Abejas/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Fosfatos de Dinucleósidos/metabolismo , Secuencia de Aminoácidos , Animales , Composición de Base , Abejas/enzimología , Biología Computacional , ADN/química , ADN/metabolismo , ADN (Citosina-5-)-Metiltransferasas/química , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Genes de Insecto , Genoma de los Insectos , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Datos de Secuencia Molecular
12.
Circulation ; 112(3): 423-31, 2005 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-16027270

RESUMEN

BACKGROUND: We previously reported that administration of elastase inhibitors reverses fatal pulmonary arterial hypertension (PAH) in rats by inducing smooth muscle cell (SMC) apoptosis. We showed in pulmonary artery (PA) organ culture that the mechanism by which elastase inhibitors induce SMC apoptosis involves repression of matrix metalloproteinase (MMP) activity and subsequent signaling through alphavbeta3-integrins and epidermal growth factor receptors (EGFRs). This suggests that blockade of these downstream effectors may also induce regression of PAH. METHODS AND RESULTS: In this study, we first showed in PA organ culture that MMP inhibition or alphavbeta3-integrin blockade with agents in clinical and preclinical use (SC-080 and cilengitide, respectively) mediates SMC apoptosis and regression of medial hypertrophy. We also documented similar results with an EGFR tyrosine kinase inhibitor. We then induced PAH in rats by injection of monocrotaline and, at day 21, began a 2-week treatment with SC-080, cilengitide, or the EGFR inhibitor PKI166. No vehicle- or cilengitide-treated animal survived beyond 2 weeks. Administration of SC-080 resulted in 44% survival at 2 weeks, and PKI166 therapy resulted in 78% and 54% survival in daily or 3-times-weekly treated animals, respectively. Four weeks after cessation of PKI166, we documented survivals of 50% and 23% in the 2 treatment groups, associated with reductions in pulmonary pressure, right ventricular hypertrophy, and abnormally muscularized distal arteries. CONCLUSIONS: We propose that selective blockade of EGFR signaling may be a novel strategy to reverse progressive, fatal PAH.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Receptores ErbB/antagonistas & inhibidores , Hipertensión Pulmonar/tratamiento farmacológico , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/efectos de los fármacos , Tirfostinos/farmacología , Animales , Hipertensión Pulmonar/mortalidad , Integrina alfaVbeta3/antagonistas & inhibidores , Masculino , Metaloproteasas/antagonistas & inhibidores , Metaloproteasas/fisiología , Miocitos del Músculo Liso/citología , Técnicas de Cultivo de Órganos , Inhibidores de Proteasas/farmacología , Quinazolinas , Ratas , Ratas Sprague-Dawley , Tirfostinos/uso terapéutico
13.
Nat Genet ; 37(3): 254-64, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15696166

RESUMEN

Transcriptional repression of methylated genes can be mediated by the methyl-CpG binding protein MeCP2. Here we show that human Brahma (Brm), a catalytic component of the SWI/SNF-related chromatin-remodeling complex, associates with MeCP2 in vivo and is functionally linked with repression. We used a number of different molecular approaches and chromatin immunoprecipitation strategies to show a unique cooperation between Brm, BAF57 and MeCP2. We show that Brm and MeCP2 assembly on chromatin occurs on methylated genes in cancer and the gene FMR1 in fragile X syndrome. These experimental findings identify a new role for SWI/SNF in gene repression by MeCP2.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Proteínas Cromosómicas no Histona/fisiología , Proteínas de Unión al ADN/fisiología , Silenciador del Gen/fisiología , Proteínas Represoras/fisiología , Transactivadores/fisiología , Factores de Transcripción/fisiología , Animales , Proteínas de Drosophila , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , Histonas/fisiología , Humanos , Proteína 2 de Unión a Metil-CpG , Ratones , Microscopía Fluorescente , Datos de Secuencia Molecular , Células 3T3 NIH , Proteínas del Tejido Nervioso/genética , Proteínas de Unión al ARN/genética
14.
J Biol Chem ; 278(41): 39830-8, 2003 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-12882977

RESUMEN

Platelet-derived growth factor (PDGF) inhibits expression of smooth muscle (SM) genes in vascular smooth muscle cells and blocks induction by arginine vasopressin (AVP). We have previously demonstrated that suppression of SM-alpha-actin by PDGF-BB is mediated in part through a Ras-dependent pathway. This study examined the role of phosphatidylinositol 3-kinase (PI3K)y and its downstream effector, Akt, in regulating SM gene expression. PDGF caused a rapid sustained activation of Akt, whereas AVP caused only a small transient increase. PDGF selectively caused a sustained stimulation of p85/p110 alpha PI3K. In contrast, p85/110 beta PI3K activity was not altered by either PDGF or AVP, whereas both agents caused a delayed activation of Class IB p101/110 gamma PI3K. Expression of a gain-of-function PI3K or myristoylated Akt (myr-Akt) mimicked the inhibitory effect of PDGF on SM-alpha-actin and SM22 alpha expression. Pretreatment with LY 294002 reversed the inhibitory effect of PDGF. Expression of myr-Akt selectively inhibited AVP-induced activation of c-Jun N-terminal kinase and p38 mitogen-activated protein kinases, which we have shown are critical for induction of these genes. Nuclear extracts from PDGF-stimulated or myr-Akt expressing cells showed reduced serum response factor binding to SM-specific CArG elements. This was associated with appearance of serum response factor in the cytoplasm. These data indicate that activation of p85/p110 alpha/Akt mediates suppression of SM gene expression by PDGF.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Arginina Vasopresina/farmacología , Becaplermina , Células Cultivadas , ADN/genética , ADN/metabolismo , Activación Enzimática/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Modelos Biológicos , Músculo Liso/efectos de los fármacos , Músculo Liso/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-sis , Ratas , Factor de Respuesta Sérica/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos
15.
EMBO J ; 22(13): 3346-55, 2003 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-12839996

RESUMEN

Herein, we define how MEKK1, a MAPK kinase kinase, regulates cell migration. MEKK1 is associated with actin fibers and focal adhesions, localizing MEKK1 to sites critical in the control of cell adhesion and migration. EGF-induced ERK1/2 activation and chemotaxis are inhibited in MEKK1-/- fibroblasts. MEKK1 deficiency causes loss of vinculin in focal adhesions of migrating cells, increased cell adhesion and impeded rear-end detachment. MEKK1 is required for activation of the cysteine protease calpain and cleavage of spectrin and talin, proteins linking focal adhesions to the cytoskeleton. Inhibition of ERK1/2 or calpain, but not of JNK, mimics MEKK1 deficiency. Therefore, MEKK1 regulates calpain-mediated substratum release of migrating fibroblasts.


Asunto(s)
Calpaína/fisiología , Movimiento Celular , Quinasa 1 de Quinasa de Quinasa MAP , Proteínas Serina-Treonina Quinasas/fisiología , Vinculina/metabolismo , Animales , Células Cultivadas , Activación Enzimática , Factor de Crecimiento Epidérmico/fisiología , Fibroblastos/citología , Fibroblastos/metabolismo , Fluorescencia , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Hidrólisis , Ratones , Papillomaviridae/fisiología , Proteínas Tirosina Quinasas/metabolismo
16.
J Heart Valve Dis ; 11(5): 726-35, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12358412

RESUMEN

BACKGROUND AND AIMS OF THE STUDY: Previous research has demonstrated that tenascin-C, an extracellular matrix protein involved in bone development and mineralization, was specifically present in calcified aortic valves, always in association with matrix metalloproteinase (MMP)-2, and was not detectable in non-calcified human aortic valves. The aim of the present study was to identify downstream targets of tenascin-C in aortic valve interstitial cells. METHODS: Subtractive hybridization was performed using sheep aortic valve interstitial cells (SAVIC) grown on a substrate of collagen plus tenascin-C, versus cells grown on type I collagen alone. RESULTS: Subtractive hybridizations revealed that nearly 70% of the clones isolated contained the sequence for thymosin beta-4, an actin-binding protein, also associated with mineralization, regulation of MMPs and inflammation. In cell culture studies, it was shown that both thymosin beta4 and thymosin beta4 sulfoxide are produced by SAVIC. When aortic valve interstitial cells were grown on a substrate of tenascin-C, thymosin beta4 expression was up-regulated. The addition of thymosin beta4 to aortic valve interstitial cell cultures resulted in a re-orientation of cytoskeletal F-actin, and induction of MMP-2. However, thymosin beta4 antisense oligonucleotide transfection did not suppress MMP-2 expression. CONCLUSION: Thymosin beta4 is up-regulated by tenascin-C, and may be involved in the primary initiation of valvular calcification.


Asunto(s)
Estenosis de la Válvula Aórtica/fisiopatología , Válvula Aórtica/fisiopatología , Calcinosis/fisiopatología , Regulación de la Expresión Génica/fisiología , Tenascina/análisis , Tenascina/fisiología , Timosina/análisis , Timosina/fisiología , Adulto , Animales , Estenosis de la Válvula Aórtica/genética , Calcinosis/genética , Bovinos , Femenino , Regulación de la Expresión Génica/genética , Humanos , Técnicas In Vitro , Masculino , Metaloproteinasas de la Matriz/análisis , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/fisiología , Ovinos , Tenascina/genética , Timosina/genética , Regulación hacia Arriba/fisiología
17.
J Biol Chem ; 277(47): 44638-50, 2002 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-12244041

RESUMEN

Important autocrine/paracrine functions for the adenine nucleotides have been proposed in several tissues. We addressed the possibility that extracellular ATP would modulate/mediate hypoxia-induced adventitial fibroblast growth. Acute hypoxia (3% O(2), 10-60 min) increased extracellular ATP concentrations in adventitial fibroblasts and in lung microvascular endothelial cells, and chronic hypoxia (3% O(2), 14-30 days) markedly attenuated the rate of extracellular ATP hydrolysis by ecto-nucleotidase(s). Exogenous ATP stimulated [(3)H]thymidine incorporation in fibroblasts as did UTP, ADPbeta, 2-methylthioadenosine triphosphate, adenosine 5'-(alpha,beta-methylene)triphosphate, and benzoylbenzoyl-ATP (2'-3'-O-(4-benzoylbenzoyl)-ATP), indicating that both P2Y and P2X purinoceptors can mediate mitogenic responses. Suramin (100 microm), Cibacron blue 3GA (100 microm), and pyridoxalphosphate-6-azophenyl-2',-4'-disulfonic acid (100 microm) as well as apyrase (5 units/ml) attenuated hypoxia- and ATP-induced and DNA synthesis, indicating activation and a functional role of purinoceptors under hypoxic conditions. ATP-induced DNA synthesis was augmented by hypoxia in an additive fashion, whereas ATP and hypoxia synergistically increased growth factor-induced DNA synthesis, again suggesting that ATP and hypoxia utilize similar signaling pathways to induce proliferation. Indeed, we found that ATP (100 microm) and hypoxia (3% O(2)) induced expression and activation of Egr-1 transcription factor, and both stimuli acted, in part, through a G(alpha)(i)/ERK1/2-dependent signaling pathway. Suramin, Cibacron blue 3GA, and apyrase attenuated hypoxia-induced ERK1/2 activation and Egr-1 expression. We conclude that hypoxia induces ATP release from endothelial cells and fibroblasts and that the activation of P2 purinoceptors is involved in the regulation of DNA synthesis by fibroblasts under hypoxic conditions.


Asunto(s)
Adenosina Trifosfato/metabolismo , Comunicación Autocrina/fisiología , Hipoxia de la Célula , Proteínas de Unión al ADN/metabolismo , Fibroblastos/fisiología , Proteínas Inmediatas-Precoces , Comunicación Paracrina/fisiología , Factores de Transcripción/metabolismo , Adenosina Trifosfatasas/metabolismo , Animales , Bovinos , División Celular/fisiología , Células Cultivadas , Medio de Cultivo Libre de Suero , ADN/biosíntesis , Proteínas de Unión al ADN/genética , Relación Dosis-Respuesta a Droga , Proteína 1 de la Respuesta de Crecimiento Precoz , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Feto/anatomía & histología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Sustancias de Crecimiento/farmacología , Humanos , Pulmón/citología , Pulmón/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Nucleótidos/metabolismo , Unión Proteica , Antagonistas del Receptor Purinérgico P2 , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA