Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 116(17): 8397-8402, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30962377

RESUMEN

Actin filaments (F-actin) are key components of sarcomeres, the basic contractile units of skeletal muscle myofibrils. A crucial step during myofibril differentiation is the sequential exchange of α-actin isoforms from smooth muscle (α-SMA) and cardiac (α-CAA) to skeletal muscle α-actin (α-SKA) that, in mice, occurs during early postnatal life. This "α-actin switch" requires the coordinated activity of actin regulators because it is vital that sarcomere structure and function are maintained during differentiation. The molecular machinery that controls the α-actin switch, however, remains enigmatic. Cyclase-associated proteins (CAP) are a family of actin regulators with largely unknown physiological functions. We here report a function for CAP2 in regulating the α-actin exchange during myofibril differentiation. This α-actin switch was delayed in systemic CAP2 mutant mice, and myofibrils remained in an undifferentiated stage at the onset of the often excessive voluntary movements in postnatal mice. The delay in the α-actin switch coincided with the onset of motor function deficits and histopathological changes including a high frequency of type IIB ring fibers. Our data suggest that subtle disturbances of postnatal F-actin remodeling are sufficient for predisposing muscle fibers to form ring fibers. Cofilin2, a putative CAP2 interaction partner, has been recently implicated in myofibril actin cytoskeleton differentiation, and the myopathies in cofilin2 and CAP2 mutant mice showed striking similarities. We therefore propose a model in which CAP2 and cofilin2 cooperate in actin regulation during myofibril differentiation.


Asunto(s)
Citoesqueleto de Actina/fisiología , Proteínas Portadoras , Diferenciación Celular , Músculo Esquelético , Miofibrillas/fisiología , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Ratones , Ratones Noqueados , Desarrollo de Músculos/genética , Desarrollo de Músculos/fisiología , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
2.
Hum Mol Genet ; 22(15): 3123-37, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23591991

RESUMEN

Mutations in ACTA2, encoding the smooth muscle cell (SMC)-specific isoform of α-actin (α-SMA), cause thoracic aortic aneurysms and dissections and occlusive vascular diseases, including early onset coronary artery disease and stroke. We have shown that occlusive arterial lesions in patients with heterozygous ACTA2 missense mutations show increased numbers of medial or neointimal SMCs. The contribution of SMC hyperplasia to these vascular diseases and the pathways responsible for linking disruption of α-SMA filaments to hyperplasia are unknown. Here, we show that the loss of Acta2 in mice recapitulates the SMC hyperplasia observed in ACTA2 mutant SMCs and determine the cellular pathways responsible for SMC hyperplasia. Acta2(-/-) mice showed increased neointimal formation following vascular injury in vivo, and SMCs explanted from these mice demonstrated increased proliferation and migration. Loss of α-SMA induced hyperplasia through focal adhesion (FA) rearrangement, FA kinase activation, re-localization of p53 from the nucleus to the cytoplasm and increased expression and ligand-independent activation of platelet-derived growth factor receptor beta (Pdgfr-ß). Disruption of α-SMA in wild-type SMCs also induced similar cellular changes. Imatinib mesylate inhibited Pdgfr-ß activation and Acta2(-/-) SMC proliferation in vitro and neointimal formation with vascular injury in vivo. Loss of α-SMA leads to SMC hyperplasia in vivo and in vitro through a mechanism involving FAK, p53 and Pdgfr-ß, supporting the hypothesis that SMC hyperplasia contributes to occlusive lesions in patients with ACTA2 missense mutations.


Asunto(s)
Actinas/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Actinas/genética , Animales , Movimiento Celular/genética , Núcleo Celular/metabolismo , Proliferación Celular , Activación Enzimática , Hiperplasia , Ratones , Ratones Noqueados , Modelos Biológicos , Fenotipo , Transporte de Proteínas , Especies Reactivas de Oxígeno/metabolismo
3.
FASEB J ; 27(2): 672-83, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23159932

RESUMEN

Elevated endothelial microparticle (MP) levels are observed in numerous diseases, increasingly supporting roles as effectors and valuable markers of vascular dysfunction. While a contractile role for the actin cytoskeleton has been implicated in vesiculation, i.e., MP production, the precise interactions and mechanisms of its constituents, ß- and γ-cytoplasmic actins, is unknown. Human cerebral microvascular endothelial cells were stimulated with known agonists, and vesiculation development was monitored by scanning electron microscopy (SEM) and flow cytometry. These data in combination provide new insight into the kinetics, patterns of vesiculating cell recruitment, and degrees of response specific to stimuli. Reorganization of ß- and γ-actins, F-actin, vinculin, and talin accompanied significant MP release. ß-Actin redistribution into basal stress fibers following stimulation was associated with increased apically situated actin-rich particulate structures, which in turn directly correlated with electron-lucent membrane protrusions observed by SEM. Y-27632 Rho-kinase inhibition abolished basal ß-actin fiber formation, minimizing apically associated actin-rich structures, significantly reducing membrane protrusions and MP release to near basal levels. Cytoskeletal protein expression and distribution varied between MPs and mother cells, as determined by Western blot. These data strongly suggest that ß-actin plays an active facilitative role in agonist-induced protuberance formation, through mechanical interactions with newly described actin-rich structures.


Asunto(s)
Actinas/fisiología , Micropartículas Derivadas de Células/fisiología , Células Endoteliales/fisiología , Actinas/ultraestructura , Amidas/farmacología , Fenómenos Biomecánicos , Calcimicina/farmacología , Línea Celular , Micropartículas Derivadas de Células/efectos de los fármacos , Micropartículas Derivadas de Células/ultraestructura , Citoesqueleto/efectos de los fármacos , Citoesqueleto/fisiología , Citoesqueleto/ultraestructura , Células Endoteliales/efectos de los fármacos , Células Endoteliales/ultraestructura , Inhibidores Enzimáticos/farmacología , Humanos , Interferón gamma/farmacología , Cinética , Lipopolisacáridos/farmacología , Microscopía Electrónica de Rastreo , Modelos Biológicos , Fragmentos de Péptidos/farmacología , Piridinas/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Quinasas Asociadas a rho/antagonistas & inhibidores
4.
J Cell Biol ; 222(7)2023 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-37204781

RESUMEN

The mechanisms that regulate the spatial sorting of nonmuscle myosins-2 (NM2) isoforms and couple them mechanically to the plasma membrane are unclear. Here we show that the cytoplasmic junctional proteins cingulin (CGN) and paracingulin (CGNL1) interact directly with NM2s through their C-terminal coiled-coil sequences. CGN binds strongly to NM2B, and CGNL1 to NM2A and NM2B. Knockout (KO), exogenous expression, and rescue experiments with WT and mutant proteins show that the NM2-binding region of CGN is required for the junctional accumulation of NM2B, ZO-1, ZO-3, and phalloidin-labeled actin filaments, and for the maintenance of tight junction membrane tortuosity and apical membrane stiffness. CGNL1 expression promotes the junctional accumulation of both NM2A and NM2B and its KO results in myosin-dependent fragmentation of adherens junction complexes. These results reveal a mechanism for the junctional localization of NM2A and NM2B and indicate that, by binding to NM2s, CGN and CGNL1 mechanically couple the actomyosin cytoskeleton to junctional protein complexes to mechanoregulate the plasma membrane.


Asunto(s)
Membrana Celular , Proteínas del Citoesqueleto , Citoesqueleto , Miosinas , Uniones Adherentes/metabolismo , Membrana Celular/metabolismo , Proteínas del Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Miosinas/metabolismo , Uniones Estrechas/metabolismo
5.
FASEB J ; 25(12): 4423-33, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21908715

RESUMEN

Cell migration plays a crucial role in numerous cellular functions, and alterations in the regulation of cell migration are required for invasive transformation of a tumor cell. While the mechanistic process of actin-based migration has been well documented, little is known as to the specific function of the nonmuscle actin isoforms in mammalian cells. Here, we present a comprehensive examination of γ-actin's role in cell migration using an RNAi approach. The partial suppression of γ-actin expression in SH-EP neuroblastoma cells resulted in a significant decrease in wound healing and transwell migration. Similarly, the knockdown of γ-actin significantly reduced speed of motility and severely affected the cell's ability to explore, which was, in part, due to a loss of cell polarity. Moreover, there was a significant increase in the size and number of paxillin-containing focal adhesions, coupled with a significant decrease in phosphorylated paxillin in γ-actin-knockdown cells. In addition, there was a significant increase in the phosphorylation of cofilin and myosin regulatory light chain, suggesting an overactivated Rho-associated kinase (ROCK) signaling pathway in γ-actin-knockdown cells. The alterations in the phosphorylation of paxillin and myosin regulatory light chain were unique to γ-actin and not ß-actin knockdown. Inhibition of the ROCK pathway with the inhibitor Y-27632 restored the ability of γ-actin-knockdown cells to migrate. This study demonstrates γ-actin as a potential upstream regulator of ROCK mediated cell migration.


Asunto(s)
Actinas/metabolismo , Movimiento Celular/fisiología , Quinasas Asociadas a rho/metabolismo , Actinas/antagonistas & inhibidores , Actinas/genética , Amidas/farmacología , Secuencia de Bases , Línea Celular , Polaridad Celular/fisiología , Adhesiones Focales/fisiología , Técnicas de Silenciamiento del Gen , Humanos , Modelos Biológicos , Cadenas Ligeras de Miosina/metabolismo , Paxillin/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/genética , Transducción de Señal , Cicatrización de Heridas/fisiología
6.
Am J Physiol Cell Physiol ; 300(6): C1356-65, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21289288

RESUMEN

Tropomyosin (Tm) is known to be an important gatekeeper of actin function. Tm isoforms are encoded by four genes, and each gene produces several variants by alternative splicing, which have been proposed to play roles in motility, proliferation, and apoptosis. Smooth muscle studies have focused on gizzard smooth muscle, where a heterodimer of Tm from the α-gene (Tmsm-α) and from the ß-gene (Tmsm-ß) is associated with contractile filaments. In this study we examined Tm in differentiated mammalian vascular smooth muscle (dVSM). Liquid chromatography-tandem mass spectrometry (LC MS/MS) analysis and Western blot screening with variant-specific antibodies revealed that at least five different Tm proteins are expressed in this tissue: Tm6 (Tmsm-α) and Tm2 from the α-gene, Tm1 (Tmsm-ß) from the ß-gene, Tm5NM1 from the γ-gene, and Tm4 from the δ-gene. Tm6 is by far most abundant in dVSM followed by Tm1, Tm2, Tm5NM1, and Tm4. Coimmunoprecipitation and coimmunofluorescence studies demonstrate that Tm1 and Tm6 coassociate with different actin isoforms and display different intracellular localizations. Using an antibody specific for cytoplasmic γ-actin, we report here the presence of a γ-actin cortical cytoskeleton in dVSM cells. Tm1 colocalizes with cortical cytoplasmic γ-actin and coprecipitates with γ-actin. Tm6, on the other hand, is located on contractile bundles. These data indicate that Tm1 and Tm6 do not form a classical heterodimer in dVSM but rather describe different functional cellular compartments.


Asunto(s)
Diferenciación Celular/fisiología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Tropomiosina/química , Tropomiosina/metabolismo , Actinas/genética , Actinas/metabolismo , Secuencia de Aminoácidos , Animales , Pollos , Hurones , Humanos , Datos de Secuencia Molecular , Miocitos del Músculo Liso/citología , Unión Proteica , Isoformas de Proteínas/genética , Alineación de Secuencia , Tropomiosina/genética
7.
Biol Reprod ; 85(5): 1025-39, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21778137

RESUMEN

In mammals, female meiosis consists of two asymmetric cell divisions, which generate a large haploid oocyte and two small polar bodies. Asymmetric partitioning of the cytoplasm results from migration of the meiotic spindle toward the cortex and requires actin filaments. However, the subcellular localization and the role of the existing two cytoplasmic actin (CYA) isoforms, beta and gamma, have not been characterized. We show that beta- and gamma-CYA are differentially distributed in the maturing oocyte from late metaphase I as well as in preimplantation embryos. Gamma-CYA is preferentially enriched in oocyte cortices and is absent from all cell-cell contact areas from metaphase II until the blastocyst stage. Beta-CYA is enriched in contractile structures, at cytokinesis, at cell-cell contacts, and around the forming blastocoel. Alteration of beta- or gamma-CYA function by isoform-specific antibody microinjection suggests that gamma-CYA holds a major and specific role in the establishment and/or maintenance of asymmetry in meiosis I and in the maintenance of overall cortical integrity. In contrast, beta- and gamma-CYA, together, appear to participate in the formation and the cortical anchorage of the second meiotic spindle in waiting for fertilization. Finally, differences in gamma-CYA expression are amongst the earliest markers of cell fate determination in development.


Asunto(s)
Actinas/fisiología , Citoplasma/fisiología , Meiosis/fisiología , Oocitos/citología , Actinas/genética , Actinas/inmunología , Animales , Anticuerpos/administración & dosificación , Anticuerpos/inmunología , Anticuerpos/farmacología , Comunicación Celular/fisiología , Diferenciación Celular/fisiología , Polaridad Celular , Células Cultivadas , Femenino , Meiosis/efectos de los fármacos , Ratones , Ratones Endogámicos , Ratones Transgénicos , Microinyecciones , Modelos Animales , Oocitos/fisiología
8.
Differentiation ; 77(4): 360-8, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19281784

RESUMEN

In the adult heart, cardiac muscle comprises the working myocardium and the conduction system (CS). The latter includes the sinoatrial node (SAN), the internodal tract or bundle (IB), the atrioventricular node (AVN), the atrioventricular bundle (AVB), the bundle branches (BB) and the peripheral Purkinje fibers (PF). Most of the information concerning the phenotypic features of CS tissue derives from the characterization of avian and rodent developing hearts; data concerning the expression of actin isoforms in adult CS cardiomyocytes are scarce. Using specific antibodies, we investigated the distribution of alpha-skeletal (alpha-SKA), alpha-cardiac (alpha-CA), alpha-smooth muscle (alpha-SMA) actin isoforms and other muscle-typical proteins in the CS of human and rat hearts at different ages. SAN and IB cardiomyocytes were characterized by the presence of alpha-SMA, alpha-CA, calponin and caldesmon, whereas alpha-SKA and vimentin were absent. Double immunofluorescence demonstrated the co-localisation of alpha-SMA and alpha-CA in I-bands of SAN cardiomyocytes. AVN, AVB, BB and PF cardiomyocytes were alpha-SMA, calponin, caldesmon and vimentin negative, and alpha-CA and alpha-SKA positive. No substantial differences in actin isoform distribution were observed in human and rat hearts, except for the presence of isolated subendocardial alpha-SMA positive cardiomyocytes co-expressing alpha-CA in the ventricular septum of the rat. Aging did not influence CS cardiomyocyte actin isoform expression profile. These findings support the concept that cardiomyocytes of SAN retain the phenotype of a developing myogenic cell throughout the entire life span.


Asunto(s)
Actinas/metabolismo , Regulación de la Expresión Génica , Sistema de Conducción Cardíaco/metabolismo , Actinas/química , Adolescente , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Animales , Células Cultivadas , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Miocitos Cardíacos/metabolismo , Isoformas de Proteínas/metabolismo , Ratas , Nodo Sinoatrial/metabolismo , Adulto Joven
9.
J Cell Mol Med ; 13(5): 896-908, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19538254

RESUMEN

Differentiation of foetal cardiomyocytes is accompanied by sequential actin isoform expression, i.e. down-regulation of the 'embryonic' alpha smooth muscle actin, followed by an up-regulation of alpha skeletal actin (alphaSKA) and a final predominant expression of alpha cardiac actin (alphaCA). Our objective was to detect whether re-expression of alphaSKA occurred during cardiomyocyte dedifferentiation, a phenomenon that has been observed in different pathologies characterized by myocardial dysfunction. Immunohistochemistry of alphaCA, alphaSKA and cardiotin was performed on left ventricle biopsies from human patients after coronary bypass surgery. Furthermore, actin isoform expression was investigated in left ventricle samples of rabbit hearts suffering from pressure- and volume-overload and in adult rabbit ventricular cardiomyocytes during dedifferentiation in vitro. Atrial goat samples up to 16 weeks of sustained atrial fibrillation (AF) were studied ultrastructurally and were immunostained for alphaCA and alphaSKA. Up-regulation of alphaSKA was observed in human ventricular cardiomyocytes showing down-regulation of alphaCA and cardiotin. A patchy re-expression pattern of alphaSKA was observed in rabbit left ventricular tissue subjected to pressure- and volume-overload. Dedifferentiating cardiomyocytes in vitro revealed a degradation of the contractile apparatus and local re-expression of alphaSKA. Comparable alphaSKA staining patterns were found in several areas of atrial goat tissue during 16 weeks of AF together with a progressive glycogen accumulation at the same time intervals. The expression of alphaSKA in adult dedifferentiating cardiomyocytes, in combination with PAS-positive glycogen and decreased cardiotin expression, offers an additional tool in the evaluation of myocardial dysfunction and indicates major changes in the contractile properties of these cells.


Asunto(s)
Actinas/metabolismo , Desdiferenciación Celular/fisiología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Actinina/metabolismo , Animales , Insuficiencia de la Válvula Aórtica/metabolismo , Insuficiencia de la Válvula Aórtica/patología , Fibrilación Atrial/metabolismo , Fibrilación Atrial/patología , Biomarcadores/metabolismo , Células Cultivadas , Enfermedad de la Arteria Coronaria/metabolismo , Enfermedad de la Arteria Coronaria/patología , Regulación hacia Abajo/fisiología , Femenino , Glucógeno/metabolismo , Cabras , Humanos , Aturdimiento Miocárdico/metabolismo , Aturdimiento Miocárdico/patología , Isoformas de Proteínas/metabolismo , Conejos , Regulación hacia Arriba/fisiología
10.
Comp Hepatol ; 8: 5, 2009 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-19602240

RESUMEN

BACKGROUND: In adult liver, the mesenchymal cells, portal fibroblasts and vascular smooth muscle cells can transdifferentiate into myofibroblasts, and are involved in portal fibrosis. Differential expression of markers, such as alpha-smooth muscle actin (ASMA), h-caldesmon and cellular retinol-binding protein-1 allows their phenotypic discrimination. The aim of our study was to explore the phenotypic evolution of the mesenchymal cells during fetal development in normal liver and in liver with portal fibrosis secondary to ductal plate malformation in a series of Meckel-Gruber syndrome, autosomal recessive polycystic kidney disease and Ivemark's syndrome. RESULTS: At the early steps of the portal tract maturation, portal mesenchymal cells expressed only ASMA. During the maturation process, these cells were found condensed around the biliary and vascular structures. At the end of maturation process, only cells around vessels expressed ASMA and cells of the artery tunica media also expressed h-caldesmon. In contrast, ASMA positive cells persisted around the abnormal biliary ducts in fibrous livers. CONCLUSION: As in adult liver, there is a phenotypic heterogeneity of the mesenchymal cells during fetal liver development. During portal tract maturation, myofibroblastic cells disappear in normal development but persist in fibrosis following ductal plate malformation.

11.
J Cell Biol ; 157(4): 657-63, 2002 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-11994316

RESUMEN

Myofibroblasts are specialized fibroblasts responsible for granulation tissue contraction and the soft tissue retractions occurring during fibrocontractive diseases. The marker of fibroblast-myofibroblast modulation is the neo expression of alpha-smooth muscle actin (alpha-SMA), the actin isoform typical of vascular smooth muscle cells that has been suggested to play an important role in myofibroblast force generation. Actin isoforms differ slightly in their NH2-terminal sequences; these conserved differences suggest different functions. When the NH2-terminal sequence of alpha-SMA Ac-EEED is delivered to cultured myofibroblast in the form of a fusion peptide (FP) with a cell penetrating sequence, it inhibits their contractile activity; moreover, upon topical administration in vivo it inhibits the contraction of rat wound granulation tissue. The NH2-terminal peptide of alpha-skeletal actin has no effect on myofibroblasts, whereas the NH2-terminal peptide of beta-cytoplasmic actin abolishes the immunofluorescence staining for this isoform without influencing alpha-SMA distribution and cell contraction. The FPs represent a new tool to better understand the specific functions of actin isoforms. Our findings support the crucial role of alpha-SMA in wound contraction. The alpha-SMA-FP will be useful for the understanding of the mechanisms of connective tissue remodeling; moreover, it furnishes the basis for a cytoskeleton-dependent preventive and/or therapeutic strategy for fibrocontractive pathological situations.


Asunto(s)
Actinas/metabolismo , Movimiento Celular/fisiología , Fibroblastos/metabolismo , Tejido de Granulación/metabolismo , Péptidos/metabolismo , Actinas/genética , Actinas/farmacología , Animales , Movimiento Celular/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Tamaño de la Célula/fisiología , Células Cultivadas , Colágeno Tipo I/biosíntesis , Colágeno Tipo I/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Tejido de Granulación/citología , Tejido de Granulación/efectos de los fármacos , Músculo Liso/metabolismo , Estructura Terciaria de Proteína , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Proteínas Recombinantes de Fusión/farmacología , Estrés Mecánico , Resistencia a la Tracción/fisiología , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología
12.
Circ Res ; 100(7): 1055-62, 2007 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-17347479

RESUMEN

We reported that smooth muscle cell (SMC) populations isolated from normal porcine coronary artery media exhibit distinct phenotypes: spindle-shaped (S) and rhomboid (R). R-SMCs are recovered in higher proportion from stent-induced intimal thickening compared with media suggesting that they participate in intimal thickening formation. Our aim was to identify a marker of R-SMCs in vitro and to explore its possible expression in vivo. S- and R-SMC protein extracts were compared by means of 2-dimensional polyacrylamide gel electrophoresis followed by tandem mass spectrometry. S100A4 was found to be predominantly expressed in R-SMC extracts. Using a monoclonal S100A4 antibody we confirmed that S100A4 is highly expressed by R-SMCs and hardly detectable in S-SMCs. S100A4 was colocalized with alpha-smooth muscle actin in stress fibers of several quiescent cells and upregulated during migration. PDGF-BB, FGF-2 or coculture with endothelial cells, which modulate S-SMCs to a R-phenotype, increased S100A4 expression in both S- and R-SMCs. Silencing of S100A4 mRNA in R-SMCs decreased cell proliferation, suggesting a functional role for this protein. In vivo S100A4 was absent in normal porcine coronary artery media, but highly expressed by SMCs of stent-induced intimal thickening. In humans, S100A4 was barely detectable in coronary artery media and markedly expressed in SMCs of atheromatous and restenotic coronary artery lesions. Our results indicate that S100A4 is a marker of porcine R-SMCs in vitro and of intimal SMCs during intimal thickening development. It is also a marker of a large population of human atheromatous and restenotic SMCs. Clarifying S100A4 function might be useful to understand the evolution of atherosclerotic and restenotic processes.


Asunto(s)
Vasos Coronarios/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas S100/metabolismo , Túnica Íntima/metabolismo , Adulto , Animales , Aterosclerosis/metabolismo , Aterosclerosis/patología , Movimiento Celular/fisiología , Proliferación Celular , Células Cultivadas , Niño , Técnicas de Cocultivo , Reestenosis Coronaria/metabolismo , Reestenosis Coronaria/patología , Vasos Coronarios/patología , Células Endoteliales/fisiología , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Proteínas Musculares/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/fisiología , Fenotipo , Proteína de Unión al Calcio S100A4 , Stents/efectos adversos , Porcinos , Distribución Tisular , Túnica Íntima/patología
13.
Front Physiol ; 9: 856, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30108507

RESUMEN

Aging induces a progressive decline in vasoconstrictor responses in central and peripheral arteries. This study investigated the hypothesis that vascular smooth muscle (VSM) contractile function declines with age in soleus muscle feed arteries (SFA). Contractile function of cannulated SFA isolated from young (4 months) and old (24 months) Fischer 344 rats was assessed by measuring constrictor responses of denuded (endothelium removed) SFA to norepinephrine (NE), phenylephrine (PE), and angiotensin II (Ang II). In addition, we investigated the role of RhoA signaling in modulation of VSM contractile function. Structural and functional characteristics of VSM cells were evaluated by fluorescence imaging and atomic force microscopy (AFM). Results indicated that constrictor responses to PE and Ang II were significantly impaired in old SFA, whereas constrictor responses to NE were preserved. In the presence of a Rho-kinase inhibitor (Y27632), constrictor responses to NE, Ang II, and PE were significantly reduced in young and old SFA. In addition, the age-group difference in constrictor responses to Ang II was eliminated. ROCK1 and ROCK2 content was similar in young and old VSM cells, whereas pROCK1 and pROCK2 were significantly elevated in old VSM cells. Aging was associated with a reduction in smooth muscle α-actin stress fibers and recruitment of proteins to cell-matrix adhesions. Old VSM cells presented an increase in integrin adhesion to the matrix and smooth muscle γ-actin fibers that was associated with increased cell stiffness. In conclusion, our results indicate that VSM contractile function declined with age in SFA. The decrement in contractile function was mediated in part by RhoA/ROCK signaling. Upregulation of pROCK in old VSM cells was not able to rescue contractility in old SFA. Collectively, these results indicate that changes at the VSM cell level play a central role in the reduced contractile function of aged SFA.

14.
Nat Commun ; 9(1): 4250, 2018 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-30315159

RESUMEN

Germline mutations in the ubiquitously expressed ACTB, which encodes ß-cytoplasmic actin (CYA), are almost exclusively associated with Baraitser-Winter Cerebrofrontofacial syndrome (BWCFF). Here, we report six patients with previously undescribed heterozygous variants clustered in the 3'-coding region of ACTB. Patients present with clinical features distinct from BWCFF, including mild developmental disability, microcephaly, and thrombocytopenia with platelet anisotropy. Using patient-derived fibroblasts, we demonstrate cohort specific changes to ß-CYA filament populations, which include the enhanced recruitment of thrombocytopenia-associated actin binding proteins (ABPs). These perturbed interactions are supported by in silico modeling and are validated in disease-relevant thrombocytes. Co-examination of actin and microtubule cytoskeleton constituents in patient-derived megakaryocytes and thrombocytes indicates that these ß-CYA mutations inhibit the final stages of platelet maturation by compromising microtubule organization. Our results define an ACTB-associated clinical syndrome with a distinct genotype-phenotype correlation and delineate molecular mechanisms underlying thrombocytopenia in this patient cohort.


Asunto(s)
Actinas/genética , Exones/genética , Trombocitopenia/genética , Actinas/metabolismo , Plaquetas/metabolismo , Células Cultivadas , Citoesqueleto/metabolismo , Femenino , Genotipo , Mutación de Línea Germinal/genética , Humanos , Masculino , Megacariocitos/metabolismo , Mutación/genética , Fenotipo , Trombocitopenia/metabolismo
15.
16.
Mol Biol Cell ; 14(6): 2508-19, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12808047

RESUMEN

Cultured myofibroblasts are characterized by stress fibers, containing alpha-smooth muscle actin (alpha-SMA) and by supermature focal adhesions (FAs), which are larger than FAs of alpha-SMA-negative fibroblasts. We have investigated the role of alpha-SMA for myofibroblast adhesion and FA maturation. Inverted centrifugation reveals two phases of initial myofibroblast attachment: during the first 2 h of plating microfilament bundles contain essentially cytoplasmic actin and myofibroblast adhesion is similar to that of alpha-SMA-negative fibroblasts. Then, myofibroblasts incorporate alpha-SMA in stress fibers, develop mature FAs and their adhesion capacity is significantly increased. When alpha-SMA expression is induced in 5 d culture by TGFbeta or low serum levels, fibroblast adhesion is further increased correlating with a "supermaturation" of FAs. Treatment of myofibroblasts with alpha-SMA fusion peptide (SMA-FP), which inhibits alpha-SMA-mediated contractile activity, reduces their adhesion to the level of alpha-SMA negative fibroblasts. With the use of flexible micropatterned substrates and EGFP-constructs we show that SMA-FP application leads to a decrease of myofibroblast contraction, shortly followed by disassembly of paxillin- and beta3 integrin-containing FAs; alpha5 integrin distribution is not affected. FRAP of beta3 integrin-EGFP demonstrates an increase of FA protein turnover following SMA-FP treatment. We conclude that the formation and stability of supermature FAs depends on a high alpha-SMA-mediated contractile activity of myofibroblast stress fibers.


Asunto(s)
Actinas/metabolismo , Fibroblastos/metabolismo , Adhesiones Focales/metabolismo , Músculo Liso/metabolismo , Animales , Citoplasma/metabolismo , Ratas
17.
Cytoskeleton (Hoboken) ; 74(4): 170-183, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28218453

RESUMEN

Actin is a major component of the cytoskeleton and is present as two isoforms in non-muscle cells: ß- and γ-cytoplasmic actin. These isoforms are strikingly conserved, differing by only four N-terminal amino acids. During spread from infected cells, vaccinia virus (VACV) particles induce localized actin nucleation that propel virus to surrounding cells and facilitate cell-to-cell spread of infection. Here we show that virus-tipped actin comets are composed of ß- and γ-actin. We employed isoform-specific siRNA knockdown to examine the role of the two isoforms in VACV-induced actin comets. Despite the high level of similarity between the actin isoforms, and their colocalization, VACV-induced actin nucleation was dependent exclusively on ß-actin. Knockdown of ß-actin led to a reduction in the release of virus from infected cells, a phenotype dependent on virus-induced Arp2/3 complex activity. We suggest that local concentrations of actin isoforms may regulate the activity of cellular actin nucleator complexes.


Asunto(s)
Actinas/metabolismo , Isoformas de Proteínas/metabolismo , Virus Vaccinia/crecimiento & desarrollo , Humanos , Virus Vaccinia/patogenicidad
18.
F1000Res ; 5: 416, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27335638

RESUMEN

Higher vertebrates express six different highly conserved actin isoforms that can be classified in three subgroups: 1) sarcomeric actins, α-skeletal (α-SKA) and α-cardiac (α-CAA), 2) smooth muscle actins (SMAs), α-SMA and γ-SMA, and 3) cytoplasmic actins (CYAs), ß-CYA and γ-CYA. The variations among isoactins, in each subgroup, are due to 3-4 amino acid differences located in their acetylated N-decapeptide sequence. The first monoclonal antibody (mAb) against an actin isoform (α-SMA) was produced and characterized in our laboratory in 1986 (Skalli  et al., 1986). We have further obtained mAbs against the 5 other isoforms. In this report, we focus on the mAb anti-α-SKA and anti-α-CAA obtained after immunization of mice with the respective acetylated N-terminal decapeptides using the Repetitive Immunizations at Multiple Sites Strategy (RIMMS). In addition to the identification of their epitope by immunoblotting, we describe the expression of the 2 sarcomeric actins in mature skeletal muscle and during muscle repair after micro-lesions. In particular, we analyze the expression of α-CAA, α-SKA and α-SMA by co-immunostaining in a time course frame during the muscle repair process. Our results indicate that a restricted myocyte population expresses α-CAA and suggest a high capacity of self-renewal in muscle cells. These antibodies may represent a helpful tool for the follow-up of muscle regeneration and pathological changes.

19.
Oncotarget ; 6(16): 14556-71, 2015 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-26008973

RESUMEN

Here we have shown that ß-cytoplasmic actin acts as a tumor suppressor, inhibiting cell growth and invasion in vitro and tumor growth in vivo. In contrast, γ-cytoplasmic actin increases the oncogenic potential via ERK1/2, p34-Arc, WAVE2, cofilin1, PP1 and other regulatory proteins. There is a positive feedback loop between γ-actin expression and ERK1/2 activation. We conclude that non-muscle actin isoforms should not be considered as merely housekeeping proteins and the ß/γ-actins ratio can be used as an oncogenic marker at least for lung and colon carcinomas. Agents that increase ß- and/or decrease γ-actin expression may be useful for anticancer therapy.


Asunto(s)
Actinas/metabolismo , Transformación Celular Neoplásica/metabolismo , Cofilina 1/metabolismo , Genes Supresores de Tumor/fisiología , Microscopía Confocal/métodos , Neoplasias/genética , Isoformas de Proteínas/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Cofilina 1/genética , Humanos , Ratones , Ratones Desnudos , Neoplasias/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Cytoskeleton (Hoboken) ; 72(6): 257-67, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26147585

RESUMEN

α-Smooth Muscle Actin (α-SMA), a widely characterized cytoskeletal protein, represents the hallmark of myofibroblast differentiation. Transforming growth factorß1 (TGFß1) stimulates α-SMA expression and incorporation into stress fibers, thus providing an increased myofibroblast contractile force that participates in tissue remodeling. We have addressed the molecular mechanism by which α-SMA is stably incorporated into stress fibers in human myofibroblasts following exposure to TGFß1. The unique N-terminal sequence AcEEED, which is critical for α-SMA incorporation into stress fibers, was used to screen for AcEEED binding proteins. Tropomyosins were identified as candidate binding proteins. We find that after TGFß1 treatment elevated levels of the Tpm1.6/7 isoforms, and to a lesser extent Tpm2.1, precede the increase in α-SMA. RNA interference experiments demonstrate that α-SMA fails to stably incorporate into stress fibers of TGFß1 treated fibroblasts depleted of Tpm1.6/7, but not other tropomyosins. This does not appear to be due to exclusive interactions between α-SMA and just the Tpm1.6/7 isoforms. We propose that an additional AcEEED binding factor may be required to generate α-SMA filaments containing just Tpm1.6/7 which result in stable incorporation of the resulting filaments into stress fibers.


Asunto(s)
Fibroblastos/metabolismo , Músculo Liso/metabolismo , Miofibroblastos/metabolismo , Isoformas de Proteínas/metabolismo , Tropomiosina/metabolismo , Humanos , Proteómica , Fibras de Estrés
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA