Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunopharmacol Immunotoxicol ; 41(3): 446-454, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31124391

RESUMEN

Context: Atherosclerosis is a chronic inflammatory disease in which the plaques were built up inside of the artery. Interleukin-8 (IL-8, CXCL8) is an inflammatory factor, known to play an important role in the development of atherosclerosis. G31P is an antagonist of the IL-8 receptor, which plays roles in vascular smooth muscle cell (VSMC) proliferation and migration. Objective: This study is to investigate the therapeutic effect of G31P on atherosclerosis through a mouse model. Materials and methods: A mouse model of atherosclerosis was generated through feeding the ApoE-/- mice with high fat diet for 12 weeks. G31P was injected subcutaneously into the mice. The levels of keratinocyte chemoattractant (KC), CXCR2, TNF-α, and IFN-γ were analyzed through ELISA. The expressions of MMP-2, MMP-9, PCNA, and Mef2a in aortic tissues were detected through RT-qPCR. In A7r5 cells, the levels of p-ERK, ROCK1, and ROCK2 were analyzed by western blot. Intracellular calcium levels were measured through Fluo-3 AM assay. Results and disccussion: G31P suppressed the abnormal lipid profile and decreased the levels of KC, MMP-2, MMP-9, PCNA, and Mef2a in a mouse model of atherosclerosis. In addition, G31P also inhibited the expressions of p-ERK, ROCK1, ROCK2, and decreased the calcium concentrations in A7r5 cells. Conclusions: These findings indicate the potential therapeutic effects of G31P in suppressing the development of atherosclerosis by antagonizing the IL-8 receptor. G31P inhibits the proliferation and migration of VSMCs through regulating the Rho-kinase, ERK, and calcium-dependent pathways.


Asunto(s)
Aorta/inmunología , Aterosclerosis/tratamiento farmacológico , Interleucina-8/farmacología , Músculo Liso Vascular/inmunología , Miocitos del Músculo Liso/inmunología , Fragmentos de Péptidos/farmacología , Placa Aterosclerótica/tratamiento farmacológico , Animales , Aorta/patología , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/patología , Citocinas/genética , Citocinas/inmunología , Modelos Animales de Enfermedad , Humanos , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/inmunología , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/inmunología , Ratones , Ratones Noqueados para ApoE , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Placa Aterosclerótica/genética , Placa Aterosclerótica/inmunología , Placa Aterosclerótica/patología , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/inmunología , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/inmunología
2.
Acta Biochim Biophys Sin (Shanghai) ; 48(6): 536-43, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27125976

RESUMEN

Physarum myosin is a Ca(2+)-binding protein and its activity is inhibited by Ca(2+) In the present study, to clarify the light chains (LCs) from the different species (Physarum and scallop) and to determine the specific Ca(2+)-regulated effects, we constructed hybrid myosins with a Physarum myosin heavy chain (Ph·HC) and Physarum and/or scallop myosin LCs, and examined Ca(2+)-mediated regulation of ATPases and motor activities. In these experiments, it was found that Ca(2+) inhibited motilities and ATPase activities of Physarum hybrid myosin with scallop regulatory light chain (ScRLC) and Physarum essential light chain (PhELC) but could not inhibit those of the Physarum hybrid myosin mutant Ph·HC/ScRLC/PhELC-3A which lacks Ca(2+)-binding ability, indicating that PhELC plays a critical role in Ca(2+)-mediated regulation of Physarum myosin. Furthermore, the effects of Ca(2+) on ATPase activities of Physarum myosin constructs are in the following order: Ph·HC/PhRLC/PhELC > Ph·HC/ScRLC/PhELC > Ph·HC/PhRLC/ScELC > Ph·HC/ScRLC/ScELC, suggesting that the presence of PhRLC and PhELC leads to the greatest Ca(2+) sensitivity of Physarum myosin. Although we did not observe the motilities of Physarum hybrid myosin Ph·HC/PhRLC/ScELC and Ph·HC/ScRLC/ScELC, our results suggest that Ca(2+)-binding to the PhELC may alter the flexibility of the regulatory domain and induce a 'closed' state, which may consequently prevent full activity and force generation.


Asunto(s)
Cadenas Pesadas de Miosina/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Pectinidae/metabolismo , Physarum/metabolismo , Secuencia de Aminoácidos , Animales , Fenómenos Biofísicos , Calcio/metabolismo , Modelos Moleculares , Movimiento , Cadenas Pesadas de Miosina/química , Cadenas Pesadas de Miosina/genética , Cadenas Ligeras de Miosina/química , Cadenas Ligeras de Miosina/genética , Pectinidae/genética , Physarum/genética , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
3.
Proc Jpn Acad Ser B Phys Biol Sci ; 92(10): 478-498, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27941307

RESUMEN

Intracellular signaling pathways include both the activation and the inhibition of biological processes. The activation of Ca2+ regulation of actin-myosin interactions was examined first, whereas it took 20 years for the author to clarify the inhibitory mode by using Physarum polycephalum, a lower eukaryote. This review describes the investigation of the inhibitory mode since 1980. The inhibitory effect of Ca2+ on myosin was detected chemically by ATPase assays and mechanically by in vitro motility assays. The Ca2+-binding ability of Physarum myosin is as high as that of scallop myosin. Ca2+ inhibits Physarum myosin, whereas it activates scallop myosin. We cloned cDNA of the myosin heavy chain and light chains to express a hybrid of Physarum and scallop myosin, and found that the Ca-binding light chain (CaLc), which belongs to an alkali light chain class, plays a major role in Ca inhibition. The role of CaLc was confirmed by mutating its EF-hand, Ca-binding structure and expressing Physarum myosin as a recombinant protein. Thus, the data obtained by classical protein purification were confirmed by the results obtained with the modern recombinant techniques. However, there are some discrepancies that remain to be solved as described in Section XII.


Asunto(s)
Actinas/metabolismo , Calcio/metabolismo , Miosinas/metabolismo , Transducción de Señal , Humanos , Unión Proteica
4.
J Pharmacol Sci ; 128(2): 78-82, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25986486

RESUMEN

The role of myosin light chain kinase (MLCK) in inducing podosomes was examined by confocal and electron microscopy. Removal of myosin from the actin core of podosomes using blebbistatin, a myosin inhibitor, resulted in the formation of smaller podosomes. Downregulation of MLCK by the transfection of MLCK small interfering RNA (siRNA) led to the failure of podosome formation. However, ML-7, an inhibitor of the kinase activity of MLCK, failed to inhibit podosome formation. Based on our previous report (Thatcher et al. J.Pharm.Sci. 116 116-127, 2011), we outlined the important role of the actin-binding activity of MLCK in producing smaller podosomes.


Asunto(s)
Quinasa de Cadena Ligera de Miosina/fisiología , Forbol 12,13-Dibutirato/farmacología , Podosomas/efectos de los fármacos , Podosomas/ultraestructura , Actinas/metabolismo , Animales , Azepinas/farmacología , Células Cultivadas , Regulación hacia Abajo , Microscopía Inmunoelectrónica , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/genética , Quinasa de Cadena Ligera de Miosina/metabolismo , Naftalenos/farmacología , Podosomas/genética , Unión Proteica , ARN Interferente Pequeño , Ratas
5.
Mol Cell Biochem ; 393(1-2): 255-63, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24792035

RESUMEN

Matrix metalloproteinases (MMP) play a pivotal role in the pathogenesis of cardiovascular diseases. Their expressions are altered in response to a variety of stimuli, including growth factors, inflammatory markers, and cytokines. In this study, we demonstrated that platelet-derived growth factor-BB (PDGF-BB) induces a dose- and time-dependent increase in MMP-2 expression in rat vascular smooth muscle cells (VSMC). Treatment with either the Rho-associated protein kinase (ROCK) inhibitor Y-27632 or suppression of ROCK-1/2 by small interfering RNA technology significantly reduced the MMP-2 expression, thus suggesting that ROCK regulates such expression. Similar results were observed when VSMC were pretreated with either U0126 or SB203580, which are selective inhibitors of extracellular signal-regulated kinase and p38 mitogen-activated protein kinase, respectively, thus suggesting that these kinases are important for the induction of MMP-2 expression by PDGF-BB. In conclusion, these results described a novel mechanism in atherosclerosis through PDGF-BB signaling in VSMC, in which MMP-2 expression is induced via extracellular signal-regulated kinases and p38 mitogen-activated protein kinase phosphorylation, as well as ROCK.


Asunto(s)
Aterosclerosis/metabolismo , Metaloproteinasa 2 de la Matriz/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Quinasas Asociadas a rho/metabolismo , Amidas/administración & dosificación , Animales , Aorta/citología , Aorta/metabolismo , Aterosclerosis/etiología , Aterosclerosis/patología , Becaplermina , Butadienos/administración & dosificación , Línea Celular , Movimiento Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Imidazoles/administración & dosificación , Metaloproteinasa 2 de la Matriz/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Nitrilos/administración & dosificación , Proteínas Proto-Oncogénicas c-sis/administración & dosificación , Proteínas Proto-Oncogénicas c-sis/metabolismo , Piridinas/administración & dosificación , Ratas , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Quinasas Asociadas a rho/antagonistas & inhibidores
6.
Biochem Biophys Res Commun ; 432(3): 488-93, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23402758

RESUMEN

Abnormal proliferation and migration of vascular smooth muscle cells (VSMC) plays an important role in vascular diseases. The Rho-associated protein kinase (ROCK) signaling pathway is now well recognized for its role in VSMC migration and proliferation. Recently, a number of studies revealed that different isoforms of ROCK have distinct functions in VSMCs. We have reported that ROCK1, rather than ROCK2, induces platelet-derived growth factor (PDGF)-BB-stimulated migration of VSMCs. In the current study, we aimed to investigate the roles of ROCK1/2 in PDGF-induced rat aorta VSMC proliferation by manipulating ROCK gene expression. The results revealed that knock-down of both ROCK1 and ROCK2 by siRNA technology decreased PDGF-BB-generated VSMC proliferation by inhibiting the expression of proliferating cell nuclear antigen (PCNA) and cyclin D1. In addition, up-regulation of ROCK1 expression through transfection, further increased the proliferation of VSMCs induced by PDGF-BB. The ERK inhibitor U0126 reduced the proliferation and expression of PCNA and cyclinD1, and ROCK1 and ROCK2 siRNA decreased the level of ERK in the nucleus. These results demonstrated that ROCK1 and ROCK2 could promote VSMC proliferation through ERK nuclear translocation, regulating the expression of PCNA and cyclin D1 protein.


Asunto(s)
Núcleo Celular/enzimología , Ciclina D1/biosíntesis , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Antígeno Nuclear de Célula en Proliferación/biosíntesis , Quinasas Asociadas a rho/fisiología , Transporte Activo de Núcleo Celular , Amidas/farmacología , Animales , Becaplermina , Proliferación Celular , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Técnicas de Silenciamiento del Gen , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Isoenzimas/fisiología , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Proteínas Proto-Oncogénicas c-sis/metabolismo , Piridinas/farmacología , Ratas , Enfermedades Vasculares/enzimología , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/genética
7.
IUBMB Life ; 65(1): 67-75, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23281038

RESUMEN

Atherosclerosis is a chronic inflammatory disease with multiple contributing factors. Hyperlipidemia is one of the major independent risks, and interleukin-8 (IL-8), as an inflammatory factor, plays an important role in the development of atherosclerosis. The aims of the study were to examine the therapeutic efficacy of G31P, an antagonist of IL-8 receptor, with a mouse model of hyperlipidemia and the potential mechanisms of G31P through the vascular smooth muscle cell (VSMC) proliferation and migration in a cell line. In vivo study: Male BALB/c mice were fed a high-fat diet for 6 months. G31P was injected subcutaneously. Blood keratinocyte chemoattractant, lipid profile, and aorta expression of inflammatory factors, matrix metalloproteinases, MMP2 and MMP9 were investigated. In vitro study: A7R5 cells were treated with IL-8 with/without G31P. Cell proliferation and migration were investigated. G31P significantly suppressed the hyperlipidermia-induced abnormal lipid profile and increased IL-8, proinflammatory factor, MMP2 and MMP9 expression. G31P also inhibited VSMC proliferation and migration both in vitro and in vivo. These findings indicate the potential therapeutic effects of G31P in preventing the development of atherosclerosis by antagonizing IL-8 receptor and decreasing the biologic activity of IL-8.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Interleucina-8/farmacología , Músculo Liso/citología , Receptores de Interleucina-8/antagonistas & inhibidores , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Interleucina-8/química , Masculino , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena de la Polimerasa , Proteínas Recombinantes/farmacología
8.
Acta Biochim Biophys Sin (Shanghai) ; 45(7): 601-6, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23665794

RESUMEN

In the present study, co-sedimentation assay, intrinsic fluorescence intensity measurement, and Mg²âº-ATPase activity analysis were carried out to investigate the direct effect of tropomyosin (TM) on unphosphorylated myosin (UM) or phosphorylated myosin (PM) in the presence or absence of caldesmon (CaD). Results showed that TM significantly decreased the sedimentation, intrinsic fluorescence intensity, and the Mg²âº-ATPase activity of PM, but not UM. In the presence of CaD, TM also significantly decreased these parameters irrespective of myosin phosphorylation, suggesting that the interaction between TM and CaD abolished the effects of TM on PM or UM and that there was an inverse interaction between TM and PM, characterized by the decreased PM sedimentation and intrinsic fluorescence intensity.


Asunto(s)
Proteínas de Unión a Calmodulina/metabolismo , Miosinas/metabolismo , Tropomiosina/metabolismo , Animales , ATPasa de Ca(2+) y Mg(2+)/metabolismo , Pollos , Fluorescencia , Fosforilación , Unión Proteica
9.
J Pharmacol Sci ; 119(1): 91-6, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22510518

RESUMEN

Myosin light-chain kinase (MLCK) is a multi-domain protein with kinase and actin-binding domains, among others. Deficiency of MLCK expression in GBaSM-4 vascular smooth muscle cells enhanced cell proliferation rate and shortened cell doubling time. Transient transfection of the MLCK-deficient cells with cDNA constructs of either wild-type MLCK or its mutant lacking the kinase activity reverted the cell proliferation rate to that of wild-type cells, whereas that of MLCK lacking the actin-binding domain maintained cell proliferation at an elevated rate similar to the MLCK-deficient cells. Thus, the actin-binding domain of MLCK seems to play a role in regulating cell proliferation.


Asunto(s)
Actinas/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Quinasa de Cadena Ligera de Miosina/genética , Actinas/genética , Animales , Línea Celular , Proliferación Celular , Regulación hacia Abajo , Cobayas , Quinasa de Cadena Ligera de Miosina/metabolismo , Fosforilación , Unión Proteica , Estructura Terciaria de Proteína , Conejos
10.
Pharmacol Res ; 63(1): 51-8, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21035550

RESUMEN

Folic acid-conjugated albumin nanospheres (FA-AN) have been developed to provide an actively targetable drug delivery system for improved drug targeting of cancer cells with reduced side effects. The nanospheres were prepared by conjugating folic acid onto the surface of albumin nanospheres using 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDAC) as a catalyst. To test the efficacy of these nanospheres as a potential delivery platform, doxorubicin-loaded albumin nanospheres (DOX-AN) and doxorubicin-loaded FA-AN (FA-DOX-AN) were prepared by entrapping DOX (an anthracycline, antibiotic drug widely used in cancer chemotherapy that works by intercalating DNA) into AN and FA-AN nanoparticles. Cell uptake of the DOX was then measured. The results show that FA-AN was incorporated into HeLa cells (tumor cells) only after 2.0h incubation, whereas HeLa cells failed to incorporate albumin nanospheres without conjugated folic acid after 4.0h incubation. When HeLa cells were treated with the DOX-AN, FA-DOX-AN nanoparticles or free DOX, cell viability decreased with increasing culture time (i.e. cell death increases with time) over a 70h period. Cell viability was always the lowest for free DOX followed by FA-DOX-AN4 and then DOX-AN. In a second set of experiments, HeLa cells washed to remove excess DOX after an initial incubation for 2h were incubated for 70h. The corresponding cell viability was slightly higher when the cells were treated with FA-DOX-AN or free DOX whilst cells treated with DOX-AN nanoparticles remained viable. The above experiments were repeated for non-cancerous, aortic smooth muscle cells (AoSMC). As expected, cell viability of the HeLa cells (with FA receptor alpha, FRα) and AoSMC cells (without FRα) decreased rapidly with time in the presence of free DOX, but treatment with FA-DOX-AN resulted in selective killing of the tumor cells. These results indicated that FA-AN may be used as a promising actively targetable drug delivery system to improve drug targeting to cancer cells.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Doxorrubicina/farmacología , Portadores de Fármacos , Ácido Fólico/metabolismo , Nanosferas , Albúmina Sérica Bovina/metabolismo , Neoplasias del Cuello Uterino/patología , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/metabolismo , Transporte Biológico , Supervivencia Celular/efectos de los fármacos , Química Farmacéutica , Doxorrubicina/química , Doxorrubicina/metabolismo , Composición de Medicamentos , Femenino , Receptor 1 de Folato/metabolismo , Ácido Fólico/análogos & derivados , Ácido Fólico/química , Células HeLa , Humanos , Luz , Microscopía Electrónica de Transmisión , Tamaño de la Partícula , Dispersión de Radiación , Albúmina Sérica Bovina/química , Solubilidad , Propiedades de Superficie , Factores de Tiempo , Neoplasias del Cuello Uterino/metabolismo
11.
Pharmacol Res ; 64(4): 410-9, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21723392

RESUMEN

In order to minimize the side effect of cancer chemotherapy, a novel galactosamine-mediated drug delivery carrier, galactosamine-conjugated albumin nanoparticles (GAL-AN), was developed for targeted liver cancer therapy. The albumin nanoparticles (AN) and doxorubicin-loaded AN (DOX-AN) were prepared by the desolvation of albumin in the presence of glutaraldehyde crosslinker. Morphological study indicated the spherical structure of these synthesized particles with an average diameter of around 200 nm. The functional ligand of galactosamine (GAL) was introduced onto the surfaces of AN and DOX-AN via carbodiimide chemistry to obtain GAL-AN and GAL-DOX-AN. Cellular uptake and kinetic studies showed that GAL-AN is able to be selectively incorporated into the HepG2 cells rather than AoSMC cells due to the existence of asialoglycoprotein receptors on HepG2 cell surface. The cytotoxicity, measured by MTT test, indicated that AN and GAL-AN are non-toxic and GAL-DOX-AN is more effective in HepG2 cell killing than that of DOX-AN. As such, our results implied that GAL-AN and GAL-DOX-AN have specific interaction with HepG2 cells via the recognition of GAL and asialoglycoprotein receptor, which renders GAL-AN a promising anticancer drug delivery carrier for liver cancer therapy.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , Sistemas de Liberación de Medicamentos , Galactosamina/química , Neoplasias Hepáticas/tratamiento farmacológico , Nanopartículas/química , Animales , Antibióticos Antineoplásicos/farmacocinética , Antibióticos Antineoplásicos/farmacología , Bovinos , Doxorrubicina/farmacocinética , Doxorrubicina/farmacología , Sistemas de Liberación de Medicamentos/métodos , Células Hep G2 , Humanos , Nanopartículas/ultraestructura , Albúmina Sérica Bovina/química
12.
Nanotechnology ; 22(24): 245101, 2011 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-21508464

RESUMEN

The potential of using actin bundles for the transport of liposomes and single cells across myosin-coated surfaces is investigated. Compared to that observed with filamentous actin, the liposome transport using actin bundles was more linear in nature and able to occur over longer distances. Bundles, but not filamentous actin, were capable of moving single cells. Cargo unloading from bundles was achieved by incubation with Triton X-100. These data suggest that actin bundling may improve the ability of the myosin motor system for nanotransport applications.


Asunto(s)
Actinas/metabolismo , Escherichia coli/citología , Escherichia coli/metabolismo , Miosina Tipo II/metabolismo , Nanoestructuras/química , Nanotecnología/métodos , Actinas/ultraestructura , Animales , Transporte Biológico , Biotinilación , Pollos , Humanos , Liposomas/ultraestructura , Microscopía Fluorescente , Nanoestructuras/ultraestructura , Conejos
13.
J Pharmacol Sci ; 115(4): 532-5, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21422730

RESUMEN

Cigarette smoking is one of the factors causing accumulation of vascular smooth muscle cells (VSMCs) in atherosclerotic plaques. Changes in cell migration toward platelet-derived growth factor BB were investigated using a Boyden chamber after 48-h preincubation of GBaSM-4 VSMCs with nicotine or nicotine-free cigarette smoke extract (CSE). A nicotine concentration of 0.1 µM maximally promoted cell migration; 0.1% CSE also promoted cell migration, while high CSE concentrations damaged GBaSM-4 cells. Fetal bovine serum (FBS) long-depletion induced decrease in migration of GBaSM-4 cells. Our results suggest that nicotine and some CSE components can induce GBaSM-4 cell migration.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/fisiología , Nicotiana/efectos adversos , Nicotina/farmacología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Humo/efectos adversos , Animales , Becaplermina , Línea Celular , Ensayos de Migración Celular/métodos , Movimiento Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Cobayas , Miocitos del Músculo Liso/efectos de los fármacos , Proteínas Proto-Oncogénicas c-sis
14.
J Pharmacol Sci ; 116(1): 116-27, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21551973

RESUMEN

Previous work has suggested that in addition to its kinase activity, myosin light chain kinase (MLCK) exhibits non-kinase properties within its N-terminus that could influence cytoskeletal organization of smooth muscle cells (A. Nakamura et al. Biochem Biophys Res Commun. 2008;369:135-143). Myosin ATPase activity measurements indicate that the 26-41 peptide of MLCK significantly decreases ATPase activity as the concentration of this peptide increases. Sliding velocity of actin-filaments on myosin and stress responses in skinned smooth muscle tissue are also inhibited. Peptide-mediated uptake and the microinjection technique in cells indicate that the peptide was necessary for actin-filament stabilization. Fluorescence resonance energy transfer analysis indicated that in the presence of MLCK, α-actin but not ß-actin remodeled during phorbol 12,13-dibutyrate (PDBu)-induced contractions. PDBu also induced podosomes in the cell. When MLCK expression was down-regulated by introduction of RNAi for MLCK by lentivirus vector into the cells, we failed to observe the podosome induction upon PDBu stimulation. Rescue experiments indicate that the non-kinase activity of MLCK plays an important role in maintaining actin stress fibers and in the PDBu-induced reorganization of actin-filaments in smooth muscle cells.


Asunto(s)
Actinas/metabolismo , Citoesqueleto/metabolismo , Contracción Muscular/efectos de los fármacos , Músculo Liso/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Forbol 12,13-Dibutirato/farmacología , Animales , Línea Celular , Extensiones de la Superficie Celular/efectos de los fármacos , Extensiones de la Superficie Celular/metabolismo , Extensiones de la Superficie Celular/ultraestructura , Pollos , Citoesqueleto/efectos de los fármacos , Inhibidores Enzimáticos/metabolismo , Silenciador del Gen , Cobayas , Técnicas In Vitro , Cinética , Proteínas de Microfilamentos/antagonistas & inhibidores , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Músculo Liso/efectos de los fármacos , Músculo Liso/ultraestructura , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/ultraestructura , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/genética , Miosinas/antagonistas & inhibidores , Fragmentos de Péptidos/metabolismo , Fosforilación/efectos de los fármacos , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , ARN Interferente Pequeño , Ratas
15.
Am J Physiol Cell Physiol ; 298(5): C1118-26, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20164381

RESUMEN

To explore the precise mechanisms of the inhibitory effects of blebbistatin, a potent inhibitor of myosin II, on smooth muscle contraction, we studied the blebbistatin effects on the mechanical properties and the structure of contractile filaments of skinned (cell membrane permeabilized) preparations from guinea pig taenia cecum. Blebbistatin at 10 microM or higher suppressed Ca(2+)-induced tension development at any given Ca(2+) concentration but had little effects on the Ca(2+)-induced myosin light chain phosphorylation. Blebbistatin also suppressed the 10 and 2.75 mM Mg(2+)-induced, "myosin light chain phosphorylation-independent" tension development at more than 10 microM. Furthermore, blebbistatin induced conformational change of smooth muscle myosin (SMM) and disrupted arrangement of SMM and thin filaments, resulting in inhibition of actin-SMM interaction irrespective of activation with Ca(2+). In addition, blebbistatin partially inhibited Mg(2+)-ATPase activity of native actomyosin from guinea pig taenia cecum at around 10 microM. These results suggested that blebbistatin suppressed skinned smooth muscle contraction through disruption of structure of SMM by the agent.


Asunto(s)
Ciego/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Contracción Muscular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Miosina Tipo II/antagonistas & inhibidores , Citoesqueleto de Actina , Actinas/metabolismo , Animales , Calcio/farmacología , Ciego/fisiología , Transferencia Resonante de Energía de Fluorescencia , Cobayas , Magnesio/farmacología , Masculino , Transporte de Proteínas , Miosinas del Músculo Liso/metabolismo
16.
Am J Physiol Heart Circ Physiol ; 298(2): H505-14, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19933418

RESUMEN

We have been searching for a mechanism to induce smooth muscle contraction that is not associated with phosphorylation of the regulatory light chain (RLC) of smooth muscle myosin (Nakamura A, Xie C, Zhang Y, Gao Y, Wang HH, Ye LH, Kishi H, Okagaki T, Yoshiyama S, Hayakawa K, Ishikawa R, Kohama K. Biochem Biophys Res Commun 369: 135-143, 2008). In this article, we report that arachidonic acid (AA) stimulates ATPase activity of unphosphorylated smooth muscle myosin with maximal stimulation (R(max)) of 6.84 +/- 0.51 relative to stimulation by the vehicle and with a half-maximal effective concentration (EC(50)) of 50.3 +/- 4.2 microM. In the presence of actin, R(max) was 1.72 +/- 0.08 and EC(50) was 26.3 +/- 2.3 microM. Our experiments with eicosanoids consisting of the AA cascade suggested that they neither stimulated nor inhibited the activity. Under conditions that did not allow RLC to be phosphorylated, AA stimulated contraction of smooth muscle tissue with an R(max) of 1.45 +/- 0.07 and an EC(50) of 27.0 +/- 4.4 microM. In addition to the ATPase activities of the myosin, AA stimulated those of heavy meromyosin, subfragment 1 (S1), S1 from which the RLC was removed, and a recombinant heavy chain consisting of the myosin head. The stimulatory effects of AA on these preparations were about twofold. The site of AA action was indicated to be the step-releasing inorganic phosphate (P(i)) from the reaction intermediate of the myosin-ADP-P(i) complex. The enhancement of P(i) release by AA was supported by computer simulation indicating that AA docked in the actin-binding cleft of the myosin motor domain. The stimulatory effect of AA was detectable with both unphosphorylated myosin and the myosin in which RLC was fully phosphorylated. The AA effect on both myosin forms was suggested to cause excess contraction such as vasospasm.


Asunto(s)
Ácido Araquidónico/farmacología , Contracción Muscular/efectos de los fármacos , Músculo Liso/enzimología , Miosinas/metabolismo , Miosinas del Músculo Liso/metabolismo , Animales , Simulación por Computador , Cobayas , Masculino , Modelos Animales , Contracción Muscular/fisiología , Músculo Liso/efectos de los fármacos , Miosinas/efectos de los fármacos , Fosfatos/metabolismo , Fosforilación , Miosinas del Músculo Liso/ultraestructura
17.
Small ; 6(3): 452-7, 2010 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-20082357

RESUMEN

The utilization of motor proteins for the movement and assembly of synthetic components is currently a goal of nanoengineering research. Application of the myosin actin motor system for nanotechnological uses has been hampered due to the low flexural rigidity of individual F-actin filaments. Here it is demonstrated how actin bundling can be used to affect the translational behavior of myosin-propelled filaments, transport molecules across a motor-patterned surface, and that the movement of bundled actin can be regulated photonically. These data suggest that actin bundling may significantly improve the applicability of the myosin motor for future nanotechnological applications.


Asunto(s)
Actinas/metabolismo , Miosinas/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/efectos de la radiación , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico/efectos de la radiación , Pollos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Luz , Nanopartículas/química , Conejos
18.
Langmuir ; 26(9): 6079-82, 2010 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-20355744

RESUMEN

The study and utilization of bionanomotors represents a rapid and progressing field of nanobiotechnology. Here, we demonstrate that poly(amidoamine) (PAMAM) dendrimers are capable of supporting heavy meromyosin dependent actin motility of similar quality to that observed using nitrocellulose, and that microcontact printing of PAMAM dendrimers can be exploited to produce tracks of active myosin motors leading to the restricted motion of actin filaments across a patterned surface. These data suggest that the use of dendrimer surfaces will increase the applicability of using protein biomolecular motors for nanotechnological applications.


Asunto(s)
Dendrímeros/química , Nanotecnología/métodos , Animales , Bovinos , Colodión/química , Electricidad , Movimiento , Miosinas/metabolismo , Propiedades de Superficie , Compuestos de Estaño/química
19.
Cell Biol Int ; 34(8): 827-35, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20388117

RESUMEN

A phosphatase was purified through a combination of ion-exchange and hydrophobic chromatography followed by native PAGE from Physarum plasmodia. Recently, we demonstrated that this phosphatase isoform has a hydrolytic activity towards the PMLC (phosphorylated light chain of Physarum myosin II) at pH 7.6. The apparent molecular mass of the purified enzyme was estimated at approximately 50 kDa by means of analytical gel filtration. The enzyme was purified 340-fold to a final phosphatase activity of 400 pkat/mg of protein. Among the phosphorylated compounds tested for hydrolytic activity at pH 7.6, the enzyme showed no activity towards nucleotides. At pH 7.6, hydrolytic activity of the enzyme against PMLC was detected; at pH 5.0, however, no hydrolytic activity towards PMLC was observed. The Km of the enzyme for PMLC was 10 microM, and the V(max) was 1.17 nkat/mg of protein. Ca(2+) (10 microM) inhibited the activity of the enzyme, and Mg(2+) (8.5 microM) activated the dephosphorylation of PMLC. Mn(2+) (1.6 microM) highly stimulated the enzyme's activity. Based on these results, we concluded that the enzyme is likely to be a phosphatase with hydrolytic activity towards PMLC.


Asunto(s)
Miosina Tipo II/metabolismo , Monoéster Fosfórico Hidrolasas/química , Physarum polycephalum/enzimología , Proteínas Protozoarias/química , Concentración de Iones de Hidrógeno , Hidrólisis , Cinética , Monoéster Fosfórico Hidrolasas/aislamiento & purificación , Monoéster Fosfórico Hidrolasas/metabolismo , Fosforilación , Isoformas de Proteínas/química , Isoformas de Proteínas/aislamiento & purificación , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Proteínas Protozoarias/aislamiento & purificación , Proteínas Protozoarias/metabolismo , Especificidad por Sustrato
20.
IUBMB Life ; 61(11): 1092-8, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19859981

RESUMEN

Myosin light-chain kinase (MLCK) comprised of N-terminal actin-binding domain, central catalytic domain, and C-terminal myosin-binding domain. It exerted not only kinase activity to phosphorylate 20 kDa regulatory light chain of smooth muscle but also exerted non-kinase activity on myosin motor and myosin ATPase activities (Nakamura et al., Biochem. Biophys. Res. Commun. 2008, 369, 135). The previous studies on the multiple MLCK functions were done using MLCK fragments. The present study reported the expression of whole MLCK molecules in Escherichia coli in a large amount. The construct in which the calmodulin (CaM) binding domain for regulating kinase activity was mutated lost the kinase activity. However, the mutant exerted non-kinase activity and inhibited both myosin motor and ATPase activities. The domain that regulated kinase activity was also shown to be involved in the Ca(2+) regulation of non-kinase activity. The deletion mutants of actin-binding domain which located at N-terminal 1-41 amino acids demonstrated that non-kinase activity was mediated through actin filaments.


Asunto(s)
Calcio/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Actinas/fisiología , Sitios de Unión , Proteínas de Unión al Calcio/metabolismo , Dominio Catalítico , Escherichia coli/enzimología , Mutación , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/efectos de los fármacos , Quinasa de Cadena Ligera de Miosina/genética , Miosinas/metabolismo , Estructura Terciaria de Proteína , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA