Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Basic Res Cardiol ; 112(1): 1, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27837311

RESUMEN

Chronic increased workload of the human heart causes ventricular hypertrophy, re-expression of the atrial essential myosin light chain (hALC-1), and improved contractile function. Although hALC-1 is an important positive inotropic regulator of the human heart, little is known about its regulation. Therefore, we investigated the role of the sex hormone 17ß-estradiol (E2) on hALC-1 gene expression, the underlying molecular mechanisms, and the impact of this regulatory process on cardiac contractile function. We showed that E2 attenuated hALC-1 expression in human atrial tissues of both sexes and in human ventricular AC16 cells. E2 induced the nuclear translocation of estrogen receptor alpha (ERα) and hALC-1 in AC16 cells, where they cooperatively regulate the transcriptional activity of hALC-1 gene promoter. E2-activated ERα required the estrogen response element (ERE) motif within the hALC-1 gene promoter to reduce its transcriptional activity (vehicle: 15.55 ± 4.80 vs. E2: 6.51 ± 3.69; ~2 fold). This inhibitory effect was potentiated in the presence of hALC-1 (vehicle: 11.13 ± 3.66 vs. E2: 2.18 ± 1.10; ~5 fold), and thus, hALC-1 acts as a co-repressor of ERα-mediated transcription. Yeast two-hybrid screening of a human heart cDNA library revealed that ERα interacts physically with hALC-1 in the presence of E2. This interaction was confirmed by Co-Immunoprecipitation and immunofluorescence in human atrium. As a further novel effect, we showed that chronic E2-treatment of adult mouse cardiomyocytes overexpressing hALC-1 resulted in reduced cell-shortening amplitude and twitching kinetics of these cells independent of Ca2+ activation levels. Together, our data showed that the expression of hALC-1 gene is, at least partly, regulated by E2/ERα, while hALC-1 acts as a co-repressor. The inotropic effect of hALC-1 overexpression in cardiomyocytes can be significantly repressed by E2.


Asunto(s)
Estradiol/metabolismo , Receptor alfa de Estrógeno/metabolismo , Regulación de la Expresión Génica/genética , Contracción Miocárdica/fisiología , Cadenas Ligeras de Miosina/biosíntesis , Animales , Western Blotting , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Miocitos Cardíacos/metabolismo , Cadenas Ligeras de Miosina/genética , Reacción en Cadena de la Polimerasa , Técnicas del Sistema de Dos Híbridos
2.
J Biol Chem ; 290(8): 4663-4676, 2015 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-25538241

RESUMEN

Cav1.3 L-type Ca(2+) channel is known to be highly expressed in neurons and neuroendocrine cells. However, we have previously demonstrated that the Cav1.3 channel is also expressed in atria and pacemaking cells in the heart. The significance of the tissue-specific expression of the channel is underpinned by our previous demonstration of atrial fibrillation in a Cav1.3 null mutant mouse model. Indeed, a recent study has confirmed the critical roles of Cav1.3 in the human heart (Baig, S. M., Koschak, A., Lieb, A., Gebhart, M., Dafinger, C., Nürnberg, G., Ali, A., Ahmad, I., Sinnegger-Brauns, M. J., Brandt, N., Engel, J., Mangoni, M. E., Farooq, M., Khan, H. U., Nürnberg, P., Striessnig, J., and Bolz, H. J. (2011) Nat. Neurosci. 14, 77-84). These studies suggest that detailed knowledge of Cav1.3 may have broad therapeutic ramifications in the treatment of cardiac arrhythmias. Here, we tested the hypothesis that there is a functional cross-talk between the Cav1.3 channel and a small conductance Ca(2+)-activated K(+) channel (SK2), which we have documented to be highly expressed in human and mouse atrial myocytes. Specifically, we tested the hypothesis that the C terminus of Cav1.3 may translocate to the nucleus where it functions as a transcriptional factor. Here, we reported for the first time that the C terminus of Cav1.3 translocates to the nucleus where it functions as a transcriptional regulator to modulate the function of Ca(2+)-activated K(+) channels in atrial myocytes. Nuclear translocation of the C-terminal domain of Cav1.3 is directly regulated by intracellular Ca(2+). Utilizing a Cav1.3 null mutant mouse model, we demonstrate that ablation of Cav1.3 results in a decrease in the protein expression of myosin light chain 2, which interacts and increases the membrane localization of SK2 channels.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Núcleo Celular/metabolismo , Regulación de la Expresión Génica/fisiología , Miocitos Cardíacos/metabolismo , Transcripción Genética/fisiología , Transporte Activo de Núcleo Celular/fisiología , Animales , Canales de Calcio Tipo L/genética , Miosinas Cardíacas/biosíntesis , Miosinas Cardíacas/genética , Núcleo Celular/genética , Atrios Cardíacos/citología , Atrios Cardíacos/metabolismo , Humanos , Ratones , Ratones Noqueados , Miocitos Cardíacos/citología , Cadenas Ligeras de Miosina/biosíntesis , Cadenas Ligeras de Miosina/genética , Estructura Terciaria de Proteína
3.
Am J Physiol Heart Circ Physiol ; 309(9): H1509-15, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26386113

RESUMEN

With the advent of high-throughput DNA sequencing, the number of identified cardiomyopathy-causing mutations has increased tremendously. As the majority of these mutations affect myofilament proteins, there is a need to understand their functional consequence on contraction. Permeabilized myofilament preparations coupled with protein exchange protocols are a common method for examining into contractile mechanics. However, producing large quantities of myofilament proteins can be time consuming and requires different approaches for each protein of interest. In the present study, we describe a unified automated method to produce troponin C, troponin T, and troponin I as well as myosin light chain 2 fused to a His6-tag followed by a tobacco etch virus (TEV) protease site. TEV protease has the advantage of a relaxed P1' cleavage site specificity, allowing for no residues left after proteolysis and preservation of the native sequence of the protein of interest. After expression in Esherichia coli, cells were lysed by sonication in imidazole-containing buffer. The His6-tagged protein was then purified using a HisTrap nickel metal affinity column, and the His6-tag was removed by His6-TEV protease digestion for 4 h at 30°C. The protease was then removed using a HisTrap column, and complex assembly was performed via column-assisted sequential desalting. This mostly automated method allows for the purification of protein in 1 day and can be adapted to most soluble proteins. It has the advantage of greatly increasing yield while reducing the time and cost of purification. Therefore, production and purification of mutant proteins can be accelerated and functional data collected in a faster, less expensive manner.


Asunto(s)
Miosinas Cardíacas/biosíntesis , Endopeptidasas , Histidina , Miofibrillas , Cadenas Ligeras de Miosina/biosíntesis , Oligopéptidos , Proteínas Recombinantes/biosíntesis , Troponina C/biosíntesis , Troponina I/biosíntesis , Troponina T/biosíntesis , Automatización , Miosinas Cardíacas/genética , Cromatografía de Afinidad , Dextranos , Escherichia coli/genética , Humanos , Cadenas Ligeras de Miosina/genética , Níquel , Proteínas Recombinantes/genética , Sefarosa , Troponina C/genética , Troponina I/genética , Troponina T/genética
4.
Am J Physiol Cell Physiol ; 301(5): C995-C1007, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21832246

RESUMEN

Skeletal muscle atrophy commonly occurs in acute and chronic disease. The expression of the muscle-specific E3 ligases atrogin-1 (MAFbx) and muscle RING finger 1 (MuRF1) is induced by atrophy stimuli such as glucocorticoids or absence of IGF-I/insulin and subsequent Akt signaling. We investigated whether glycogen synthase kinase-3ß (GSK-3ß), a downstream molecule in IGF-I/Akt signaling, is required for basal and atrophy stimulus-induced expression of atrogin-1 and MuRF1, and myofibrillar protein loss in C(2)C(12) skeletal myotubes. Abrogation of basal IGF-I signaling, using LY294002, resulted in a prominent induction of atrogin-1 and MuRF1 mRNA and was accompanied by a loss of myosin heavy chain fast (MyHC-f) and myosin light chains 1 (MyLC-1) and -3 (MyLC-3). The synthetic glucocorticoid dexamethasone (Dex) also induced the expression of both atrogenes and likewise resulted in the loss of myosin protein abundance. Genetic ablation of GSK-3ß using small interfering RNA resulted in specific sparing of MyHC-f, MyLC-1, and MyLC-3 protein levels after Dex treatment or impaired IGF-I/Akt signaling. Interestingly, loss of endogenous GSK-3ß suppressed both basal and atrophy stimulus-induced atrogin-1 and MuRF1 expression, whereas pharmacological GSK-3ß inhibition, using CHIR99021 or LiCl, only reduced atrogin-1 mRNA levels in response to LY294002 or Dex. In conclusion, our data reveal that myotube atrophy and myofibrillar protein loss are GSK-3ß dependent, and demonstrate for the first time that basal and atrophy stimulus-induced atrogin-1 mRNA expression requires GSK-3ß enzymatic activity, whereas MuRF1 expression depends solely on the physical presence of GSK-3ß.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , Músculo Esquelético/enzimología , Atrofia Muscular/enzimología , Mioblastos/enzimología , Animales , Línea Celular , Cromonas/farmacología , Dexametasona/farmacología , Inhibidores Enzimáticos/farmacología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Cloruro de Litio/farmacología , Ratones , Morfolinas/farmacología , Proteínas Musculares/biosíntesis , Músculo Esquelético/efectos de los fármacos , Atrofia Muscular/tratamiento farmacológico , Mioblastos/efectos de los fármacos , Mioblastos/fisiología , Cadenas Pesadas de Miosina/biosíntesis , Cadenas Ligeras de Miosina/biosíntesis , Piridinas/farmacología , Pirimidinas/farmacología , ARN Interferente Pequeño/metabolismo , Proteínas Ligasas SKP Cullina F-box/biosíntesis , Transducción de Señal/efectos de los fármacos , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas/biosíntesis
5.
J Periodontal Res ; 45(5): 618-25, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20546111

RESUMEN

BACKGROUND AND OBJECTIVE: The junctional epithelium attaches to the tooth enamel at the dentogingival junction. The attachment mechanisms of the junctional epithelium have been studied histologically, but the molecular functions of the junctional epithelium have not been elucidated. The aim of this study was to perform a comprehensive analysis of gene expression in the junctional epithelium and to search for specific genetic markers of the junctional epithelium. MATERIAL AND METHODS: A comprehensive analysis of genes expressed in the mouse junctional epithelium and oral gingival epithelium was performed using laser microdissection and microarray analysis. To extract high-quality RNA from these tissues, we made frozen sections using a modified film method. Confirmation of the differential expression of selected genes was performed by quantitative real-time PCR and immunohistochemistry. RESULTS: The modified method produced RNA of sufficient quality for microarray analysis. The result of microarray analysis showed that 841 genes were up-regulated in the junctional epithelium compared with the oral gingival epithelium, and five were increased more than 50-fold in the junctional epithelium. These five genes were secretory leukocyte protease inhibitor (Slpi), keratin 17 (Krt17), annexin A1 (Anxa1), myosin light peptide 6 (Myl6) and endoplasmic reticulum protein 29 (Erp29). In particular, Slpi expression in the junctional epithelium was approximately 100-fold higher than in the oral gingival epithelium by real-time PCR. Additionally, immunohistochemistry indicated that the Slpi protein is highly expressed in the junctional epithelium. CONCLUSION: We developed a method for generating fresh-frozen tissue sections suitable for extraction of good-quality RNA. We determined that Slpi is characteristically expressed in the junctional epithelium. Our results provide a substantial advance in the analysis of gene expression in the junctional epithelium.


Asunto(s)
Inserción Epitelial/metabolismo , Perfilación de la Expresión Génica/métodos , Inhibidor Secretorio de Peptidasas Leucocitarias/biosíntesis , Animales , Anexina A1/biosíntesis , Anexina A1/genética , Retículo Endoplásmico , Inserción Epitelial/enzimología , Secciones por Congelación , Encía/metabolismo , Proteínas de Choque Térmico/biosíntesis , Proteínas de Choque Térmico/genética , Queratina-17/biosíntesis , Queratina-17/genética , Láseres de Gas , Ratones , Microdisección/métodos , Cadenas Ligeras de Miosina/biosíntesis , Cadenas Ligeras de Miosina/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Inhibidor Secretorio de Peptidasas Leucocitarias/genética
6.
Am J Physiol Heart Circ Physiol ; 297(1): H191-9, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19429828

RESUMEN

The participation of nonmuscle myosin in force maintenance is controversial. Furthermore, its regulation is difficult to examine in a cellular context, as the light chains of smooth muscle and nonmuscle myosin comigrate under native and denaturing electrophoresis techniques. Therefore, the regulatory light chains of smooth muscle myosin (SM-RLC) and nonmuscle myosin (NM-RLC) were purified, and these proteins were resolved by isoelectric focusing. Using this method, intact mouse aortic smooth muscle homogenates demonstrated four distinct RLC isoelectric variants. These spots were identified as phosphorylated NM-RLC (most acidic), nonphosphorylated NM-RLC, phosphorylated SM-RLC, and nonphosphorylated SM-RLC (most basic). During smooth muscle activation, NM-RLC phosphorylation increased. During depolarization, the increase in NM-RLC phosphorylation was unaffected by inhibition of either Rho kinase or PKC. However, inhibition of Rho kinase blocked the angiotensin II-induced increase in NM-RLC phosphorylation. Additionally, force for angiotensin II stimulation of aortic smooth muscle from heterozygous nonmuscle myosin IIB knockout mice was significantly less than that of wild-type littermates, suggesting that, in smooth muscle, activation of nonmuscle myosin is important for force maintenance. The data also demonstrate that, in smooth muscle, the activation of nonmuscle myosin is regulated by Ca(2+)-calmodulin-activated myosin light chain kinase during depolarization and a Rho kinase-dependent pathway during agonist stimulation.


Asunto(s)
Músculo Liso/fisiología , Miosinas/metabolismo , Miosina Tipo IIB no Muscular/metabolismo , Secuencia de Aminoácidos , Angiotensina II/farmacología , Animales , Western Blotting , Pollos , Electroforesis en Gel Bidimensional , Regulación de la Expresión Génica/fisiología , Espectrometría de Masas , Ratones , Ratones Transgénicos , Contracción Muscular/fisiología , Músculo Liso/efectos de los fármacos , Cadenas Ligeras de Miosina/biosíntesis , Cadenas Ligeras de Miosina/química , Cadenas Ligeras de Miosina/metabolismo , Miosinas/química , Miosinas/genética , Miosina Tipo IIB no Muscular/química , Miosina Tipo IIB no Muscular/genética , Fosforilación/efectos de los fármacos , Proteínas Recombinantes , Quinasas Asociadas a rho/antagonistas & inhibidores
7.
Circ Res ; 100(3): 354-62, 2007 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-17234970

RESUMEN

The sinoatrial node, which resides at the junction of the right atrium and the superior caval vein, contains specialized myocardial cells that initiate the heart beat. Despite this fundamental role in heart function, the embryonic origin and mechanisms of localized formation of the sinoatrial node have not been defined. Here we show that subsequent to the formation of the Nkx2-5-positive heart tube, cells bordering the inflow tract of the heart tube give rise to the Nkx2-5-negative myocardial cells of the sinoatrial node and the sinus horns. Using genetic models, we show that as the myocardium of the heart tube matures, Nkx2-5 suppresses pacemaker channel gene Hcn4 and T-box transcription factor gene Tbx3, thereby enforcing a progressive confinement of their expression to the forming Nkx2-5-negative sinoatrial node and sinus horns. Thus, Nkx2-5 is essential for establishing a gene expression border between the atrium and sinoatrial node. Tbx3 was found to suppress chamber differentiation, providing an additional mechanism by which the Tbx3-positive sinoatrial node is shielded from differentiating into atrial myocardium. Pitx2c-deficient fetuses form sinoatrial nodes with indistinguishable molecular signatures at both the right and left sinuatrial junction, indicating that Pitx2c functions within the left/right pathway to suppress a default program for sinuatrial node formation on the left. Our molecular pathway provides a mechanism for how pacemaker activity becomes progressively relegated to the most recently added components of the venous pole of the heart and, ultimately, to the junction of the right atrium and superior caval vein.


Asunto(s)
Tipificación del Cuerpo/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Atrios Cardíacos/embriología , Ventrículos Cardíacos/embriología , Proteínas de Homeodominio/fisiología , Canales Iónicos/biosíntesis , Nodo Sinoatrial/embriología , Proteínas de Dominio T Box/fisiología , Factores de Transcripción/fisiología , Animales , Factor Natriurético Atrial , Biomarcadores , Tipificación del Cuerpo/genética , Miosinas Cardíacas/biosíntesis , Miosinas Cardíacas/genética , Conexinas/biosíntesis , Conexinas/genética , Canales Catiónicos Regulados por Nucleótidos Cíclicos , Regulación del Desarrollo de la Expresión Génica/genética , Genes Reporteros , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Imagenología Tridimensional , Hibridación in Situ , Canales Iónicos/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Miocardio/metabolismo , Cadenas Ligeras de Miosina/biosíntesis , Cadenas Ligeras de Miosina/genética , Péptido Natriurético Tipo-C/biosíntesis , Péptido Natriurético Tipo-C/genética , Precursores de Proteínas/biosíntesis , Precursores de Proteínas/genética , Proteínas Recombinantes de Fusión/fisiología , Nodo Sinoatrial/citología , Proteínas de Dominio T Box/biosíntesis , Proteínas de Dominio T Box/genética , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Troponina I/biosíntesis , Troponina I/genética , Proteína alfa-5 de Unión Comunicante , Proteína del Homeodomínio PITX2
8.
PLoS One ; 14(4): e0214982, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30964931

RESUMEN

Disuse induces adaptations in skeletal muscle, which lead to muscle deterioration. Hindlimb-unloading (HU) is a well-established model to investigate cellular mechanisms responsible for disuse-induced skeletal muscle dysfunction. In myosin heavy chain (MHC) type IIB fibers HU induces a reduction in contraction speed (Vo) and a reduction in the relative myosin light chain 3f (MLC3f) protein content compared with myosin light chain 1f (MLC1f) protein. This study tested the hypothesis that increasing the relative MLC3f protein content via rAd-MLC3f vector delivery would attenuate the HU-induced decline in Vo in single MHC type IIB fibers. Fischer-344 rats were randomly assigned to one of three groups: control, HU for 7 days, and HU for 7 days plus rAd-MLC3f. The semimembranosus muscles were injected with rAd-MLC3f (3.75 x 1011-5 x 1011 ifu/ml) at four days after the initiation of HU. In single MHC type IIB fibers the relative MLC3f content decreased by 25% (12.00±0.60% to 9.06±0.66%) and Vo was reduced by 29% (3.22±0.14fl/s vs. 2.27±0.08fl/s) with HU compared to the control group. The rAd-MLC3f injection resulted in an increase in the relative MLC3f content (12.26±1.19%) and a concomitant increase in Vo (2.90±0.15fl/s) of MHC type IIB fibers. A positive relationship was observed between the percent of MLC3f content and Vo. Maximal isometric force and specific tension were reduced with HU by 49% (741.45±44.24µN to 379.09±23.77µN) and 33% (97.58±4.25kN/m2 to 65.05±2.71kN/m2), respectively compared to the control group. The rAd-MLC3f injection did not change the HU-induced decline in force or specific tension. Collectively, these results indicate that rAd-MLC3f injection rescues hindlimb unloading-induced decline in Vo in MHC type IIB single muscle fibers.


Asunto(s)
Adaptación Fisiológica , Contracción Muscular , Fibras Musculares Esqueléticas/metabolismo , Trastornos Musculares Atróficos/prevención & control , Cadenas Ligeras de Miosina/biosíntesis , Adenoviridae , Animales , Vectores Genéticos , Suspensión Trasera , Masculino , Fibras Musculares Esqueléticas/patología , Trastornos Musculares Atróficos/genética , Trastornos Musculares Atróficos/metabolismo , Cadenas Ligeras de Miosina/genética , Ratas , Ratas Endogámicas F344 , Transducción Genética
9.
Gene Expr Patterns ; 7(7): 738-45, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17638597

RESUMEN

Mrf4 (Myf6) is a member of the basic helix-loop-helix (bHLH) myogenic regulatory transcription factor (MRF) family, which also contains Myod, Myf5 and myogenin. Mrf4 is implicated in commitment of amniote cells to skeletal myogenesis and is also abundantly expressed in many adult muscle fibres. The specific role of Mrf4 is unclear both because mrf4 null mice are viable, suggesting redundancy with other MRFs, and because of genetic interactions at the complex mrf4/myf5 locus. We report the cloning and expression of an mrf4 gene from zebrafish, Danio rerio, which shows conservation of linkage to myf5. Mrf4 mRNA accumulates in a subset of terminally differentiated muscle fibres in parallel with myosin protein in the trunk and fin. Although most, possibly all, trunk muscle expresses mrf4, the level of mRNA is dynamically regulated. No expression is detected in muscle precursor cell populations prior to myosin accumulation. Moreover, mrf4 expression is not detected in head muscles, at least at early stages. As fish mature, mrf4 expression is pronounced in the region of slow muscle fibres.


Asunto(s)
Factores Reguladores Miogénicos/biosíntesis , Factores Reguladores Miogénicos/fisiología , Secuencia de Aminoácidos , Amnios/citología , Animales , Miosinas Cardíacas/biosíntesis , Miosinas Cardíacas/fisiología , Regulación de la Expresión Génica , Modelos Biológicos , Datos de Secuencia Molecular , Músculos/metabolismo , Factor 5 Regulador Miogénico/metabolismo , Miogenina/metabolismo , Cadenas Ligeras de Miosina/biosíntesis , Cadenas Ligeras de Miosina/fisiología , Filogenia , Homología de Secuencia de Aminoácido , Distribución Tisular , Pez Cebra
10.
J Clin Invest ; 98(2): 467-73, 1996 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-8755658

RESUMEN

Most of the patients with congenital heart diseases express the atrial myosin light chain 1 (ALC-1) in the right ventricle. We investigated the functional consequences of ALC-1 expression on the myosin cycling kinetics in the intact sarcomeric structure using multicellular demembranated fibers ("skinned fibers") from the right ventricular infundibulum of patients with Tetralogy of Fallot (TOF), double outlet right ventricle (DORV), and infundibular pulmonary stenosis (IPS), Force-velocity relation was analyzed by the constant-load technique at maximal Ca2+ activation (pCa 4.5). Half-time of tension development (t1/2) was investigated by monitoring contraction initiation upon photolytic release of ATP from caged-ATP in rigor. The patients investigated here expressed between 0 and 27% ALC-1. There was a statistically significant correlation between ALC-l and maximal shortening velocity (Vmax) which rose 1.87-fold from 1.2 muscle length per second (ML/s) to 2.25 ML/s in a normal (0% ALC-1) and diseased (19.9% ALC-1) ventricle. Half-time of tension development decreased 1.85-fold with increasing ALC-1 expression (t1/2) was 0.252 s and 0.136 s at 2 and 18.4% ALC-1, respectively). We conclude that the expression of ALC-1 in the human heart modulates cross-bridge cycling kinetics accelerating shortening velocity and isometric tension production.


Asunto(s)
Presión Sanguínea , Cardiopatías Congénitas/fisiopatología , Corazón/fisiopatología , Contracción Miocárdica , Cadenas Ligeras de Miosina/fisiología , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/metabolismo , Calcio/farmacología , Niño , Preescolar , Femenino , Corazón/efectos de los fármacos , Corazón/fisiología , Cardiopatías Congénitas/cirugía , Ventrículos Cardíacos , Homeostasis , Humanos , Lactante , Cinética , Masculino , Persona de Mediana Edad , Músculo Liso Vascular/fisiopatología , Contracción Miocárdica/efectos de los fármacos , Cadenas Ligeras de Miosina/biosíntesis , Arteria Pulmonar/fisiopatología , Estenosis de la Válvula Pulmonar/fisiopatología , Valores de Referencia , Tetralogía de Fallot/fisiopatología
11.
J Card Fail ; 13(8): 680-6, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17923362

RESUMEN

BACKGROUND: Dilated cardiomyopathy is a naturally occurring disease in humans and dogs. Human studies have shown increased levels of myosin heavy chain (MHC)-beta in failing ventricles and the left atria (LA) and of ventricular light chain (VLC)-2 in the right atria in dilated cardiomyopathy. METHODS AND RESULTS: This study evaluates the levels of MHC-beta in all heart chambers in prolonged canine right ventricular pacing. In addition, we determined whether levels of VLC2 were altered in these hearts. Failing hearts demonstrated significantly increased levels of MHC-beta in the right atria, right atrial appendage, LA, left atrial appendage (LAA), and right ventricle compared with controls. Significant levels of VLC2 were detected in the right atria of paced hearts. Differences in MHC-beta expression were observed between the LA and the LAA of paced and control dogs. MHC-beta expression was significantly greater in the LA of paced and control dogs compared with their respective LAA. CONCLUSIONS: The cardiac myosin isoform shifts in this study were similar to those observed in end-stage human heart failure and more severe than those reported in less prolonged pacing models, supporting the use of this model for further study of end-stage human heart failure. The observation of consistent differences between sampling sites, especially LA versus LAA, indicates the need for rigorous sampling consistency in future studies.


Asunto(s)
Función del Atrio Derecho/fisiología , Cardiomiopatías/metabolismo , Cadenas Pesadas de Miosina/biosíntesis , Cadenas Ligeras de Miosina/biosíntesis , Miosina Tipo IIB no Muscular/biosíntesis , Animales , Cardiomiopatías/genética , Perros , Regulación de la Expresión Génica/fisiología , Cadenas Pesadas de Miosina/genética , Cadenas Ligeras de Miosina/genética , Miosina Tipo IIB no Muscular/genética
12.
Mol Cell Biol ; 16(7): 3909-22, 1996 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-8668209

RESUMEN

The fast alkali myosin light chain 1f/3f (MLC1f/3f) gene is developmentally regulated, muscle specific, and preferentially expressed in fast-twitch fibers. A transgene containing an MLC1f promoter plus a downstream enhancer replicates this pattern of expression in transgenic mice. Unexpectedly, this transgene is also expressed in a striking (approximately 100-fold) rostrocaudal gradient in axial muscles (reviewed by J. R. Sanes, M. J. Donoghue, M. C. Wallace, and J. P. Merlie, Cold Spring Harbor Symp. Quant. Biol. 57:451-460, 1992). Here, we analyzed the expression of mutated transgenes to map sites necessary for muscle-specific, fiber-type-selective, and axially graded expression. We show that two E boxes (myogenic factor binding sites), a homeodomain (hox) protein binding site, and an MEF2 site, which are clustered in an approximately 170-bp core enhancer, are all necessary for maximal transgene activity in muscle but not for fiber-type- or position-dependent expression. A distinct region within the core enhancer promotes selective expression of the transgene in fast-twitch muscles. Sequences that flank the core enhancer are also necessary for high-level activity in transgenic mice but have little influence on activity in transfected cells, suggesting the presence of regions resembling matrix attachment sites. Truncations of the MLC1f promoter affected position-dependent expression of the transgene, revealing distinct regions that repress transgene activity in neck muscles and promote differential expression among intercostal muscles. Thus, the whole-body gradient of expression displayed by the complete transgene may reflect the integrated activities of discrete elements that regulate expression in subsets of muscles. Finally, we show that transgene activity is not significantly affected by deletion or overexpression of the myoD gene, suggesting that intermuscular differences in myogenic factor levels do not affect patterns of transgene expression. Together, our results provide evidence for at least nine distinct sites that exert major effects on the levels and patterns of MLC1f expression in adult muscles.


Asunto(s)
Elementos de Facilitación Genéticos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Cadenas Ligeras de Miosina/biosíntesis , Regiones Promotoras Genéticas , Secuencias Reguladoras de Ácidos Nucleicos , Animales , Secuencia de Bases , Cloranfenicol O-Acetiltransferasa/biosíntesis , Cruzamientos Genéticos , Huella de ADN , Cartilla de ADN , Desoxirribonucleasa I , Embrión de Mamíferos/citología , Embrión de Mamíferos/fisiología , Desarrollo Embrionario y Fetal , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Datos de Secuencia Molecular , Músculo Esquelético/embriología , Mutagénesis Sitio-Dirigida , Cadenas Ligeras de Miosina/genética , Especificidad de Órganos , Reacción en Cadena de la Polimerasa , Proteínas Recombinantes/biosíntesis , Transfección
13.
Brain Res Bull ; 74(6): 439-51, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17920452

RESUMEN

Movement of glutamate receptors in neurons likely involves direct and indirect association of receptor subunits with microtubule- and actin-based motor proteins. We have previously shown that myosin II regulatory light chain (RLC) binds directly to subunits of the NMDA-type glutamate receptor (NR), suggesting that NMDA receptors are closely associated with a myosin II motor complex. Using a polyclonal antibody predicted to recognize all RLC isoforms previously described in rodent brain, we report the expression of RLC and the NR1 subunit in cortex, hippocampus and cerebellum of postnatal day 0 (P0) and adult mouse. Although myosin RLC was not exclusively localized with NR1 by immunohistochemistry, co-staining was striking in the neuronal soma of deep cortical neurons and Purkinje neurons of the cerebellum which showed a punctate, perinuclear pattern of immunoreactivity. These neuronal populations were identified using a monoclonal antibody directed against a nuclear-specific, transcriptional repressor, chicken ovalbumin upstream promoter-transcription factor (COUP-TF)-interacting protein 2 (CTIP2). Co-expression of NR1 and a myosin II motor was validated using an isoform specific anti-nonmuscle myosin II-B heavy chain (NMHC II-B) antibody. Our findings support the idea that there is regional heterogeneity in the molecular composition of the NMDA receptor-associated cytoskeleton, and suggest that NR subunits may be associated with an actin-based, myosin II-B motor within the endomembrane system of some neuronal populations. Differential staining patterns observed with light and heavy chain antibodies, however, suggest that there is also heterogeneity in the composition of myosin II complexes in brain.


Asunto(s)
Encéfalo/metabolismo , Cadenas Ligeras de Miosina/biosíntesis , Miosina Tipo II/biosíntesis , Receptores de N-Metil-D-Aspartato/biosíntesis , Animales , Animales Recién Nacidos , Western Blotting , Femenino , Inmunohistoquímica , Ratones
14.
Mol Biol Cell ; 14(7): 2781-92, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12857864

RESUMEN

The function of the GTPase Rac1, a molecular switch transducing intracellular signals from growth factors, in differentiation of a specific cell type during early embryogenesis has not been investigated. To address the question, we used embryonic stem (ES) cells differentiated into cardiomyocytes, a model that faithfully recapitulates early stages of cardiogenesis. Overexpression in ES cells of a constitutively active Rac (RacV12) but not of an active mutant (RacL61D38), which does not activate the NADPH oxydase generating ROS, prevented MEF2C expression and severely compromised cardiac cell differentiation. This resulted in poor expression of ventricular myosin light chain 2 (MLC2v) and its lack of insertion into sarcomeres. Thus ES-derived cardiomyocytes featured impaired myofibrillogenesis and contractility. Overexpression of MEF2C or addition of catalase in the culture medium rescued the phenotype of racV12 cells. In contrast, RacV12 specifically expressed in ES-derived ventricular cells improved the propensity of cardioblasts to differentiate into beating cardiomyocytes. This was attributed to both a facilitation of myofibrillogenesis and a prolongation in their proliferation. The dominant negative mutant RacN17 early or lately expressed in ES-derived cells prevented myofibrillogenesis and in turn beating of cardiomyocytes. We thus suggest a stage-dependent function of the GTPase during early embryogenesis.


Asunto(s)
Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Corazón/embriología , Miocitos Cardíacos/metabolismo , Células Madre/citología , Proteína de Unión al GTP rac1/metabolismo , Animales , Células Cultivadas , Regulación hacia Abajo , Factores de Transcripción MEF2 , Ratones , Desarrollo de Músculos/genética , Desarrollo de Músculos/fisiología , Mutación , Miocitos Cardíacos/citología , Factores Reguladores Miogénicos/metabolismo , Cadenas Ligeras de Miosina/biosíntesis , NADPH Oxidasas/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Sarcómeros/metabolismo , Células Madre/metabolismo , Proteína de Unión al GTP rac1/genética
15.
Circulation ; 112(9 Suppl): I123-8, 2005 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-16159803

RESUMEN

BACKGROUND: Angiogenesis is enhanced after transplantation of vascular endothelial growth factor (VEGF)-expressing cells into a myocardial scar. Insulin-like growth factor I (IGF-I) may induce hypertrophy and inhibit apoptosis. We evaluated the effect of cell-based IGF-I and VEGF multigene therapy on left ventricular (LV) function, cell survival, and apoptosis after bone marrow cell (BMC) transplantation. METHODS AND RESULTS: Female Lewis rats underwent left anterior descending ligation 3 weeks before transplantation with male donor BMC, BMC transfected with VEGF (BMC+VEGF), IGF-I (BMC+IGF-I), VEGF and IGF-I (BMC+VEGF+IGF-I), or medium without cells (control) (n=4 per group x 5 groups x 4 time points). Three days and 1, 2, and 4 weeks after transplantation, VEGF and IGF-I expression was quantitated by real-time polymerase chain reaction, cell survival by polymerase chain reaction for sry2, apoptosis by TUNEL staining, LV function by echocardiography and myosin heavy chain, and light chain and troponin I by Western blot. One week after transplantation, IGF-I expression in the scar and border zone was greatest in BMC+IGF-I and BMC+VEGF+IGF-I rats (P<0.05). VEGF expression in the scar and border zone was greatest in BMC+VEGF and BMC+VEGF+IGF-I hearts (P<0.05). Transplanted cell survival was lowest in BMC, intermediate in BMC+VEGF and BMC+IGF-I, and greatest in BMC+VEGF+IGF-I (P<0.05). Apoptotic indices were significantly reduced in BMC+VEGF+IGF-I, BMC+VEGF, and BMC+IGF-I (P<0.05). Two and 4 weeks after transplantation, LV ejection fraction was lowest in control, intermediate in BMC, BMC+VEGF, and BMC+IGF-I, and greatest in BMC+VEGF+IGF-I (P<0.05). CONCLUSIONS: Transplantation of VEGF- and IGF-I-expressing BMC reduced apoptosis, maximized transplanted cell survival, and enhanced LV function. Multimodal cell-based gene therapy may maximize the benefits of cell transplantation.


Asunto(s)
Trasplante de Médula Ósea , Terapia Genética , Factor I del Crecimiento Similar a la Insulina/fisiología , Trasplante de Células Madre Mesenquimatosas , Infarto del Miocardio/terapia , Factor A de Crecimiento Endotelial Vascular/fisiología , Disfunción Ventricular Izquierda/terapia , Función Ventricular Izquierda , Animales , Apoptosis , Células de la Médula Ósea/citología , Miosinas Cardíacas/biosíntesis , Miosinas Cardíacas/genética , Supervivencia Celular , Células Cultivadas/trasplante , Cicatriz/patología , Femenino , Supervivencia de Injerto , Factor I del Crecimiento Similar a la Insulina/biosíntesis , Factor I del Crecimiento Similar a la Insulina/genética , Masculino , Infarto del Miocardio/complicaciones , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Cadenas Pesadas de Miosina/biosíntesis , Cadenas Pesadas de Miosina/genética , Cadenas Ligeras de Miosina/biosíntesis , Cadenas Ligeras de Miosina/genética , Neovascularización Fisiológica , Ratas , Ratas Endogámicas Lew , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/fisiología , Volumen Sistólico , Transfección , Troponina I/biosíntesis , Troponina I/genética , Ultrasonografía , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/genética , Disfunción Ventricular Izquierda/diagnóstico por imagen , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/fisiopatología
16.
Circ Res ; 92(6): 609-16, 2003 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-12623879

RESUMEN

Despite the advantages of reversibly altering cardiac transgene expression, the number of successful studies with inducible cardiac-specific transgene expression remains limited. The utility of the current system is hampered by the large number of lines needed before a nonleaky inducible line is isolated and by the use of a heterologous virus-based minimal promoter in the responder line. We developed an efficient, experimentally flexible system that enables us to reversibly affect both abundant and nonabundant cardiomyocyte proteins. The use of bacterial-codon-based transactivators led to aberrant splicing, whereas other more efficient transactivators, by themselves, caused disease when expressed in the heart. The redesign of the system focused on developing stable transactivator-expressing lines in which expression was driven by the mouse alpha-myosin heavy chain promoter. A minimal responder locus was derived from the same promoter, in which the GATA sites and thyroid responsive elements responsible for robust cardiac specific expression were ablated, leading to an attenuated promoter that could be inducibly controlled. In all cases, whether activated or not, expression mimicked that of the parental promoter. By use of this system, an inducible expression of an abundant contractile protein, the atrial isoform of essential myosin light chain 1, and a powerful biological effector, glycogen synthase kinase-3beta (GSK-3beta), were obtained. Subsequently, we tested the hypothesis that GSK-3beta expression could reverse a preexisting hypertrophy. Inducible expression of GSK-3beta could both attenuate a hypertrophic response and partially reverse a pressure-overload-induced hypertrophy. The system appears to be robust and can be used to temporally control high levels of cardiac-specific transgene expression.


Asunto(s)
Ingeniería Genética/métodos , Miocardio/metabolismo , Cadenas Pesadas de Miosina/genética , Regiones Promotoras Genéticas , Activación Transcripcional , Animales , Cardiomegalia/etiología , ADN Complementario/metabolismo , Glucógeno Sintasa Quinasa 3/biosíntesis , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Proteína Vmw65 de Virus del Herpes Simple/análisis , Proteína Vmw65 de Virus del Herpes Simple/biosíntesis , Proteína Vmw65 de Virus del Herpes Simple/genética , Ratones , Ratones Transgénicos , Cadenas Ligeras de Miosina/biosíntesis , Cadenas Ligeras de Miosina/genética , Empalme del ARN , Sarcómeros/metabolismo , Eliminación de Secuencia , Tetraciclina/farmacología , Transgenes
17.
Nucleic Acids Res ; 32(4): 1577-83, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15004244

RESUMEN

The BOB.1/OBF.1 protein is a B-cell-specific coactivator of the Oct1 and Oct2 transcription factors. It is involved in mediating the transcriptional activity of the Oct proteins. However, animals deficient for BOB.1/OBF.1 showed virtually normal expression of genes that contain octamer motifs in their regulatory regions. To identify new genes that are regulated by BOB.1/OBF.1, we took advantage of a previously described cell system. RNAs differentially expressed in a BOB.1/OBF.1-deficient pre-B cell line and a derivative of this cell line expressing a hormone dependent BOB.1/OBF.1-estrogen receptor (BobER) fusion protein were isolated. Using the cDNA representational difference analysis method we could identify myosin light chain 1 atrial (MLC1A) isoform as a gene regulated by BOB.1/OBF.1. MLC1A was so far unknown to be expressed in tissues other than muscle. Here we demonstrate that MLC1A is indeed expressed in mouse pre-B cells. Analysis of the expressed mRNA revealed an alternative 5' promoter element and an alternative splice product, which had not yet been described for the murine gene. Cotransfection experiments with reporter constructs driven by the MLC1A promoter suggest that the regulation by BOB.1/OBF.1 is indirect. Consistent with this conclusion is the observation that transcriptional induction of the endogenous MLC1A gene by BOB.1/OBF.1 requires de novo protein synthesis.


Asunto(s)
Linfocitos B/metabolismo , Cadenas Ligeras de Miosina/genética , Células Madre/metabolismo , Transactivadores/fisiología , Animales , Linfocitos B/citología , Secuencia de Bases , Línea Celular , Regulación de la Expresión Génica , Ratones , Datos de Secuencia Molecular , Músculos/metabolismo , Cadenas Ligeras de Miosina/biosíntesis , ARN Mensajero/metabolismo , Activación Transcripcional
18.
Mol Med Rep ; 14(5): 4293-4300, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27748856

RESUMEN

The role of myosin light chain II (MLC­II) in cellular differentiation of rat mandibular condylar chondrocytes (MCCs) induced by cyclical uniaxial compressive stress (CUCS) remains unclear. In the current study, a four­point bending system was used to apply CUCS to primary cultured MCCs from rats. It was identified that CUCS stimulated features of cellular differentiation including morphological alterations, cytoskeleton rearrangement and overproduction of proteoglycans. Furthermore, CUCS promoted runt­related transcription factor­2 (RUNX2) expression at mRNA (P<0.01) and protein levels (P<0.05) and elevated alkaline phosphatase (ALP) activity (P<0.01), which are both markers of osteogenic differentiation. Under conditions of stress, western blotting indicated that the ratio of phosphorylated MLC­II to total MLC­II was increased significantly (P<0.05). Silencing MLC­II by RNA interference reduced ALP activity (P<0.01), and eliminated RUNX2 mRNA expression (P<0.01). Addition of the MLC kinase inhibitor, ML­7, reduced the CUCS­associated upregulation of RUNX2 expression (P<0.01) and ALP activity (P<0.01). The data indicated that CUCS promoted cellular differentiation of rat primary MCCs, and this was suggested to be via the phosphorylation of MLC­II.


Asunto(s)
Miosinas Cardíacas/genética , Condrocitos/citología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/biosíntesis , Cóndilo Mandibular/crecimiento & desarrollo , Cadenas Ligeras de Miosina/genética , Osteogénesis/genética , Fosfatasa Alcalina/antagonistas & inhibidores , Fosfatasa Alcalina/genética , Animales , Azepinas/administración & dosificación , Miosinas Cardíacas/antagonistas & inhibidores , Miosinas Cardíacas/biosíntesis , Diferenciación Celular/genética , Condrocitos/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Citoesqueleto/genética , Citoesqueleto/metabolismo , Regulación del Desarrollo de la Expresión Génica , Cóndilo Mandibular/citología , Cóndilo Mandibular/metabolismo , Cadenas Ligeras de Miosina/antagonistas & inhibidores , Cadenas Ligeras de Miosina/biosíntesis , Naftalenos/administración & dosificación , Fosforilación , Presión , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas
19.
Biol Trace Elem Res ; 173(1): 116-25, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26779623

RESUMEN

Selenium (Se), a nutritionally essential trace element, is associated with health and disease. Selenoprotein T (SelT) was identified as a redoxin protein with a selenocystein, localizing in the endoplasmic reticulum. The myosin light chain kinase (MLCK) and myosin light chain (MLC) play key roles in the contraction process of smooth muscle. The present study was to detect the effect and mechanism of SelT on the contraction process of gastric smooth muscle. The WT rats were fed with different Se concentration diets, and Se and Ca(2+) concentrations were detected in the gastric smooth muscle. Western blot and qPCR were performed to determine SelT, CaM, MLCK, and MLC expressions. MLCK activity was measured by identifying the rates of [γ-32P]ATP incorporated into the MLC. The results showed Se and Ca(2+) concentrations were enhanced with Se intake in gastric smooth muscle tissues. With increasing Se, SelT, CaM, MLCK and MLC expressions increased, and MLCK and MLC activation improved in gastric smooth muscle tissue. The SelT RNA interference experiments showed that Ca(2+) release, MLCK activation, and MLC phosphorylation were regulated by SelT. Se affected the gastric smooth muscle constriction by regulating Ca(2+) release, MLCK activation, and MLC phosphorylation through SelT. Se plays a major role in regulating the contraction processes of gastric smooth muscle with the SelT.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Mucosa Gástrica/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Contracción Muscular/efectos de los fármacos , Músculo Liso/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Selenio/farmacología , Selenoproteínas/biosíntesis , Animales , Activación Enzimática/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Cadenas Ligeras de Miosina/biosíntesis , Ratas , Ratas Wistar
20.
Biochim Biophys Acta ; 1519(1-2): 139-42, 2001 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-11406284

RESUMEN

We have isolated MLC1(F) (tMLC1(F)), MLC3(F) (tMLC3(F)) and skeletal actin cDNAs from the teleost Oncorhynchus mykiss. Sequence analysis indicates that tMLC1(F) and tMLC3(F) are not produced from differentially spliced mRNAs as reported in avians and rodents but are encoded by different genes. Results from RNase protection analysis showed that the corresponding transcripts are expressed in fast skeletal muscles. Whole-mount in situ hybridisation revealed distinct expression patterns of the myosin alkali light chains and skeletal actin genes during skeletal muscle development in the embryo.


Asunto(s)
Actinas/genética , Regulación del Desarrollo de la Expresión Génica , Músculo Esquelético/metabolismo , Cadenas Ligeras de Miosina/genética , Oncorhynchus mykiss/genética , Actinas/biosíntesis , Secuencia de Aminoácidos , Animales , ADN Complementario/biosíntesis , ADN Complementario/aislamiento & purificación , Embrión no Mamífero/metabolismo , Datos de Secuencia Molecular , Músculo Esquelético/embriología , Cadenas Ligeras de Miosina/biosíntesis , Oncorhynchus mykiss/embriología , Oncorhynchus mykiss/metabolismo , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA