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











Base de datos
Intervalo de año de publicación
1.
JACC Heart Fail ; 6(8): 619-632, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30071950

RESUMEN

The number of persons with heart failure has continued to rise over the last several years. Approximately one-half of those living with heart failure have heart failure with preserved ejection fraction, but critical unsolved questions remain across the spectrum of basic, translational, clinical, and population research in heart failure with preserved ejection fraction. In this study, the authors summarize existing knowledge, persistent controversies, and gaps in evidence with regard to the understanding of heart failure with preserved ejection fraction. Our analysis is based on an expert panel discussion "Think Tank" meeting that included representatives from academia, the National Institutes of Health, the U.S. Food and Drug Administration, the Centers for Medicare & Medicaid Services, and industry.


Asunto(s)
Insuficiencia Cardíaca/fisiopatología , Volumen Sistólico , Medicina Basada en la Evidencia , Testimonio de Experto , Insuficiencia Cardíaca/clasificación , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/terapia , Humanos
2.
Circulation ; 135(4): 352-362, 2017 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-27986651

RESUMEN

BACKGROUND: Alirocumab, a monoclonal antibody to proprotein convertase subtilisin/kexin type 9 (PCSK9), lowers plasma low-density lipoprotein (LDL) cholesterol and apolipoprotein B100 (apoB). Although studies in mice and cells have identified increased hepatic LDL receptors as the basis for LDL lowering by PCSK9 inhibitors, there have been no human studies characterizing the effects of PCSK9 inhibitors on lipoprotein metabolism. In particular, it is not known whether inhibition of PCSK9 has any effects on very low-density lipoprotein or intermediate-density lipoprotein (IDL) metabolism. Inhibition of PCSK9 also results in reductions of plasma lipoprotein (a) levels. The regulation of plasma Lp(a) levels, including the role of LDL receptors in the clearance of Lp(a), is poorly defined, and no mechanistic studies of the Lp(a) lowering by alirocumab in humans have been published to date. METHODS: Eighteen (10 F, 8 mol/L) participants completed a placebo-controlled, 2-period study. They received 2 doses of placebo, 2 weeks apart, followed by 5 doses of 150 mg of alirocumab, 2 weeks apart. At the end of each period, fractional clearance rates (FCRs) and production rates (PRs) of apoB and apo(a) were determined. In 10 participants, postprandial triglycerides and apoB48 levels were measured. RESULTS: Alirocumab reduced ultracentrifugally isolated LDL-C by 55.1%, LDL-apoB by 56.3%, and plasma Lp(a) by 18.7%. The fall in LDL-apoB was caused by an 80.4% increase in LDL-apoB FCR and a 23.9% reduction in LDL-apoB PR. The latter was due to a 46.1% increase in IDL-apoB FCR coupled with a 27.2% decrease in conversion of IDL to LDL. The FCR of apo(a) tended to increase (24.6%) without any change in apo(a) PR. Alirocumab had no effects on FCRs or PRs of very low-density lipoproteins-apoB and very low-density lipoproteins triglycerides or on postprandial plasma triglycerides or apoB48 concentrations. CONCLUSIONS: Alirocumab decreased LDL-C and LDL-apoB by increasing IDL- and LDL-apoB FCRs and decreasing LDL-apoB PR. These results are consistent with increases in LDL receptors available to clear IDL and LDL from blood during PCSK9 inhibition. The increase in apo(a) FCR during alirocumab treatment suggests that increased LDL receptors may also play a role in the reduction of plasma Lp(a). CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: NCT01959971.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Lipoproteínas VLDL/metabolismo , Inhibidores de PCSK9 , Adolescente , Adulto , Anciano , Anticuerpos Monoclonales Humanizados , Femenino , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
3.
J Am Heart Assoc ; 5(6)2016 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-27287699

RESUMEN

BACKGROUND: Alirocumab undergoes target-mediated clearance via binding of proprotein convertase subtilisin/kexin type 9 (PCSK9). Statins increase PCSK9 levels; the effects of nonstatin lipid-lowering therapies are unclear. Every-4-weeks dosing of alirocumab may be appropriate for some patients in absence of background statin but is not yet approved. METHODS AND RESULTS: Low-density lipoprotein cholesterol (LDL-C), PCSK9, and alirocumab levels were assessed in subjects (LDL-C >130 mg/dL, n=24/group) after a 4-week run-in taking oral ezetimibe, fenofibrate, or ezetimibe placebo, when alirocumab 150 mg every 4 weeks (days 1, 29, and 57) was added. Maximal mean LDL-C reductions from day -1 baseline (prealirocumab) occurred on day 71 in all groups: alirocumab plus placebo, 47.4%; alirocumab plus ezetimibe, 56.6%; and alirocumab plus fenofibrate, 54.3%. LDL-C reductions were sustained through day 85 with alirocumab plus placebo (47.0%); the duration of effect was slightly diminished at day 85 versus day 71 with ezetimibe (49.6%) or fenofibrate combinations (43.2%). Free PCSK9 concentrations were lowest at day 71 in all groups, then increased over time; by day 85, free PCSK9 concentrations were higher, and alirocumab levels lower, with alirocumab plus fenofibrate, and to a lesser extent alirocumab plus ezetimibe, versus alirocumab plus placebo. CONCLUSIONS: Alirocumab 150 mg every 4 weeks produced maximal LDL-C reductions of 47% in combination with placebo and 54% to 57% in combination with ezetimibe or fenofibrate. The oral lipid-lowering therapies appear to increase PCSK9 levels, leading to increased alirocumab clearance. Although the duration of effect was modestly diminished with alirocumab plus ezetimibe/fenofibrate versus placebo, the effect was less than observed in trials with background statins, and it would not preclude the use of alirocumab every 4 weeks in patients taking these nonstatin lipid-lowering therapies concomitantly. CLINICAL TRIAL REGISTRATION: URL: http://www.Clinicaltrials.gov. Unique identifier: NCT01723735.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticolesterolemiantes/farmacología , LDL-Colesterol/metabolismo , Fenofibrato/administración & dosificación , Hipercolesterolemia/tratamiento farmacológico , Proproteína Convertasa 9/metabolismo , Administración Oral , Adolescente , Adulto , Anciano , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Anticolesterolemiantes/administración & dosificación , Anticolesterolemiantes/farmacocinética , Anticolesterolemiantes/uso terapéutico , Método Doble Ciego , Quimioterapia Combinada , Ezetimiba/administración & dosificación , Femenino , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Hipercolesterolemia/sangre , Hipolipemiantes/administración & dosificación , Masculino , Persona de Mediana Edad , Proproteína Convertasa 9/efectos de los fármacos , Adulto Joven
4.
Am J Physiol Cell Physiol ; 307(9): C878-92, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25143350

RESUMEN

Microvascular stability and regulation of capillary tonus are regulated by pericytes and their interactions with endothelial cells (EC). While the RhoA/Rho kinase (ROCK) pathway has been implicated in modulation of pericyte contractility, in part via regulation of the myosin light chain phosphatase (MLCP), the mechanisms linking Rho GTPase activity with actomyosin-based contraction and the cytoskeleton are equivocal. Recently, the myosin phosphatase-RhoA-interacting protein (MRIP) was shown to mediate the RhoA/ROCK-directed MLCP inactivation in vascular smooth muscle. Here we report that MRIP directly interacts with the ß-actin-specific capping protein ßcap73. Furthermore, manipulation of MRIP expression influences pericyte contractility, with MRIP silencing inducing cytoskeletal remodeling and cellular hypertrophy. MRIP knockdown induces a repositioning of ßcap73 from the leading edge to stress fibers; thus MRIP-silenced pericytes increase F-actin-driven cell spreading twofold. These hypertrophied and cytoskeleton-enriched pericytes demonstrate a 2.2-fold increase in contractility upon MRIP knockdown when cells are plated on a deformable substrate. In turn, silencing pericyte MRIP significantly affects EC cycle progression and angiogenic activation. When MRIP-silenced pericytes are cocultured with capillary EC, there is a 2.0-fold increase in EC cycle entry. Furthermore, in three-dimensional models of injury and repair, silencing pericyte MRIP results in a 1.6-fold elevation of total tube area due to EC network formation and increased angiogenic sprouting. The pivotal role of MRIP expression in governing pericyte contractile phenotype and endothelial growth should lend important new insights into how chemomechanical signaling pathways control the "angiogenic switch" and pathological angiogenic induction.


Asunto(s)
Células Endoteliales/fisiología , Endotelio Vascular/citología , Neovascularización Fisiológica , Pericitos/metabolismo , Pericitos/ultraestructura , Proteínas de Capping de la Actina/metabolismo , Animales , Células COS , Bovinos , Ciclo Celular , Tamaño de la Célula , Células Cultivadas , Chlorocebus aethiops , Citoesqueleto/ultraestructura , Células Endoteliales/citología , Humanos , Ratones , Proteínas de Microfilamentos/antagonistas & inhibidores , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Células 3T3 NIH , Pericitos/citología , Interferencia de ARN , Quinasas Asociadas a rho/metabolismo
5.
Atherosclerosis ; 230(1): 52-60, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23958252

RESUMEN

OBJECTIVE: This study evaluated the effects of anacetrapib (ANA) on lipids and safety when administered as monotherapy or in combination with atorvastatin (ATV) in Japanese patients with dyslipidemia. METHODS: Patients (n = 407) were randomized equally to 1 of 10 groups: placebo, ATV 10 mg, ANA 10, 40, 100, or 300 mg once daily, and the same ANA doses in combination with ATV 10 mg. Patients were treated with study medication for 8 weeks and followed for an additional 8 weeks, during which ANA was switched to placebo. RESULTS: For the placebo and ANA monotherapy groups (10, 40, 100, and 300 mg), least squares mean percent changes from baseline at Week 8 for low-density lipoprotein cholesterol (LDL-C) calculated by the Friedewald equation were 3%, -12%, -27%, -32%, and -32%, respectively, and for high-density lipoprotein-cholesterol (HDL-C) were 1%, 56%, 116%, 134%, and 159%, respectively (p < 0.001 vs. placebo for all doses). All ANA doses co-administered with ATV 10 mg produced significantly greater LDL-C reductions and HDL-C increases compared with ATV 10 mg monotherapy. ANA was well tolerated, and dose-dependent relationships for adverse events were not observed across treatment groups. Changes from baseline in blood pressure and electrolytes were not significantly different between the active and control treatment groups. CONCLUSION: ANA, as monotherapy or co-administered with ATV, produced significant reductions in LDL-C and increases in HDL-C. ANA was generally well tolerated in Japanese patients with dyslipidemia.


Asunto(s)
Proteínas de Transferencia de Ésteres de Colesterol/sangre , Dislipidemias/tratamiento farmacológico , Oxazolidinonas/uso terapéutico , Adulto , Anciano , Anticolesterolemiantes/uso terapéutico , Atorvastatina , Proteínas de Transferencia de Ésteres de Colesterol/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Dislipidemias/sangre , Femenino , Regulación de la Expresión Génica , Ácidos Heptanoicos/uso terapéutico , Humanos , Japón , Masculino , Persona de Mediana Edad , Seguridad del Paciente , Pirroles/uso terapéutico , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
6.
J Biol Chem ; 288(34): 24972-83, 2013 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-23831687

RESUMEN

NO, via its second messenger cGMP, activates protein kinase GI (PKGI) to induce vascular smooth muscle cell relaxation. The mechanisms by which PKGI kinase activity regulates cardiovascular function remain incompletely understood. Therefore, to identify novel protein kinase G substrates in vascular cells, a λ phage coronary artery smooth muscle cell library was constructed and screened for phosphorylation by PKGI. The screen identified steroid-sensitive gene 1 (SSG1), which harbors several predicted PKGI phosphorylation sites. We observed direct and cGMP-regulated interaction between PKGI and SSG1. In cultured vascular smooth muscle cells, both the NO donor S-nitrosocysteine and atrial natriuretic peptide induced SSG1 phosphorylation, and mutation of SSG1 at each of the two predicted PKGI phosphorylation sites completely abolished its basal phosphorylation by PKGI. We detected high SSG1 expression in cardiovascular tissues. Finally, we found that activation of PKGI with cGMP regulated SSG1 intracellular distribution.


Asunto(s)
Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , GMP Cíclico/metabolismo , Regulación de la Expresión Génica/fisiología , Glicoproteínas/biosíntesis , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Supresoras de Tumor/biosíntesis , Células Cultivadas , GMP Cíclico/genética , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/genética , Cisteína/análogos & derivados , Cisteína/farmacología , Proteínas de la Matriz Extracelular , Regulación de la Expresión Génica/efectos de los fármacos , Glicoproteínas/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Donantes de Óxido Nítrico/farmacología , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , S-Nitrosotioles/farmacología , Proteínas Supresoras de Tumor/genética
7.
BMC Genet ; 11: 22, 2010 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-20398283

RESUMEN

BACKGROUND: Sequencing of the human genome has identified numerous chromosome copy number additions and subtractions that include stable partial gene duplications and pseudogenes that when not properly annotated can interfere with genetic analysis. As an example of this problem, an evolutionary chromosome event in the primate ancestral chromosome 18 produced a partial duplication and inversion of rho-associated protein kinase 1 (ROCK1 -18q11.1, 33 exons) in the subtelomeric region of the p arm of chromosome 18 detectable only in humans. ROCK1 and the partial gene copy, which the gene databases also currently call ROCK1, include non-unique single nucleotide polymorphisms (SNPs). RESULTS: Here, we characterize this partial gene copy of the human ROCK1, termed Little ROCK, located at 18p11.32. Little ROCK includes five exons, four of which share 99% identity with the terminal four exons of ROCK1 and one of which is unique to Little ROCK. In human while ROCK1 is expressed in many organs, Little ROCK expression is restricted to vascular smooth muscle cell (VSMC) lines and organs rich in smooth muscle. The single nucleotide polymorphism database (dbSNP) lists multiple variants contained in the region shared by ROCK1 and Little ROCK. Using gene and cDNA sequence analysis we clarified the origins of two non-synonymous SNPs annotated in the genome to actually be fixed differences between the ROCK1 and the Little ROCK gene sequences. Two additional coding SNPs were valid polymorphisms selectively within Little ROCK. Little ROCK-Green Fluorescent fusion proteins were highly unstable and degraded by the ubiquitin-proteasome system in vitro. CONCLUSION: In this report we have characterized Little ROCK (ROCK1P1), a human expressed pseudogene derived from partial duplication of ROCK1. The large number of pseudogenes in the human genome creates significant genetic diversity. Our findings emphasize the importance of taking into consideration pseudogenes in all candidate gene and genome-wide association studies, as well as the need for complete annotation of human pseudogenome.


Asunto(s)
Músculo Liso/metabolismo , Polimorfismo de Nucleótido Simple , Seudogenes , Quinasas Asociadas a rho/genética , Secuencia de Bases , Cromosomas Humanos Par 18 , Duplicación de Gen , Humanos , Datos de Secuencia Molecular , Músculo Liso Vascular/metabolismo , Alineación de Secuencia , Quinasas Asociadas a rho/metabolismo
8.
Circ Res ; 104(4): 531-40, 2009 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-19131646

RESUMEN

Abnormal vascular smooth muscle cell (VSMC) contraction plays an important role in vascular diseases. The RhoA/ROCK signaling pathway is now well recognized to mediate vascular smooth muscle contraction in response to vasoconstrictors by inhibiting myosin phosphatase (MLCP) activity and increasing myosin light chain phosphorylation. Two ROCK isoforms, ROCK1 and ROCK2, are expressed in many tissues, yet the isoform-specific roles of ROCK1 and ROCK2 in vascular smooth muscle and the mechanism of ROCK-mediated regulation of MLCP are not well understood. In this study, ROCK2, but not ROCK1, bound directly to the myosin binding subunit of MLCP, yet both ROCK isoforms regulated MLCP and myosin light chain phosphorylation. Despite that both ROCK1 and ROCK2 regulated MLCP, the ROCK isoforms had distinct and opposing effects on VSMC morphology and ROCK2, but not ROCK1, had a predominant role in VSMC contractility. These data support that although the ROCK isoforms both regulate MLCP and myosin light chain phosphorylation through different mechanisms, they have distinct roles in VSMC function.


Asunto(s)
Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Cadenas Ligeras de Miosina/metabolismo , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Vasoconstricción , Quinasas Asociadas a rho/metabolismo , Animales , Sitios de Unión , Línea Celular , Forma de la Célula , Células Cultivadas , Humanos , Isoenzimas , Lisofosfolípidos/metabolismo , Fosforilación , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo , Transfección , Quinasas Asociadas a rho/genética
9.
J Biol Chem ; 283(47): 32860-9, 2008 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-18782776

RESUMEN

Nitric oxide and nitrovasodilators induce vascular smooth muscle cell relaxation in part by cGMP-dependent protein kinase I (PKG-Ialpha)-mediated activation of myosin phosphatase (MLCP). Mechanistically it has been proposed that protein-protein interactions between the N-terminal leucine zipper (LZ) domain of PKG-Ialpha ((PKG-Ialpha(1-59)) and the LZ and/or coiled coil (CC) domain of the myosin binding subunit (MBS) of MLCP are localized in the C terminus of MBS. Although recent studies have supported these interactions, the critical amino acids responsible for these interactions have not been identified. Here we present structural and biophysical data identifying that the LZ domain of PKG-Ialpha(1-59) interacts with a well defined 42-residue CC motif (MBS(CT42)) within the C terminus of MBS. Using glutathione S-transferase pulldown experiments, chemical cross-linking, size exclusion chromatography, circular dichroism, and isothermal titration calorimetry we identified a weak dimer-dimer interaction between PKG-Ialpha(1-59) and this C-terminal CC domain of MBS. The K(d) of this non-covalent complex is 178.0+/-1.5 microm. Furthermore our (1)H-(15)N heteronuclear single quantum correlation NMR data illustrate that this interaction is mediated by several PKG-Ialpha residues that are on the a, d, e, and g hydrophobic and electrostatic interface of the C-terminal heptad layers 2, 4, and 5 of PKG-Ialpha. Taken together these data support a role for the LZ domain of PKG-Ialpha and the CC domain of MBS in this requisite contractile complex.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/química , Cadenas Ligeras de Miosina/química , Miosinas/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Dicroismo Circular , Reactivos de Enlaces Cruzados/química , Proteína Quinasa Dependiente de GMP Cíclico Tipo I , Cinética , Leucina/química , Leucina Zippers , Espectroscopía de Resonancia Magnética , Datos de Secuencia Molecular , Monoéster Fosfórico Hidrolasas/química , Unión Proteica , Estructura Terciaria de Proteína
10.
Proc Natl Acad Sci U S A ; 105(18): 6702-7, 2008 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-18448676

RESUMEN

Hypertension, a major cardiovascular risk factor and cause of mortality worldwide, is thought to arise from primary renal abnormalities. However, the etiology of most cases of hypertension remains unexplained. Vascular tone, an important determinant of blood pressure, is regulated by nitric oxide, which causes vascular relaxation by increasing intracellular cGMP and activating cGMP-dependent protein kinase I (PKGI). Here we show that mice with a selective mutation in the N-terminal protein interaction domain of PKGIalpha display inherited vascular smooth muscle cell abnormalities of contraction, abnormal relaxation of large and resistance blood vessels, and increased systemic blood pressure. Renal function studies and responses to changes in dietary sodium in the PKGIalpha mutant mice are normal. These data reveal that PKGIalpha is required for normal VSMC physiology and support the idea that high blood pressure can arise from a primary abnormality of vascular smooth muscle cell contractile regulation, suggesting a new approach to the diagnosis and therapy of hypertension and cardiovascular diseases.


Asunto(s)
Hipertensión/fisiopatología , Músculo Liso Vascular/fisiopatología , Aldosterona/sangre , Animales , Proteína Quinasa Dependiente de GMP Cíclico Tipo I , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Dieta , Hipertensión/enzimología , Ratones , Músculo Liso Vascular/enzimología , Resistencia Vascular , Sistema Vasomotor/enzimología , Proteína de Unión al GTP rhoA/metabolismo
11.
J Cell Biochem ; 103(4): 1158-70, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-17661354

RESUMEN

Vascular smooth muscle cell contractile state is the primary determinant of blood vessel tone. Vascular smooth muscle cell contractility is directly related to the phosphorylation of myosin light chains (MLCs), which in turn is tightly regulated by the opposing activities of myosin light chain kinase (MLCK) and myosin phosphatase. Myosin phosphatase is the principal enzyme that dephosphorylates MLCs leading to relaxation. Myosin phosphatase is regulated by both vasoconstrictors that inhibit its activity to cause MLC phosphorylation and contraction, and vasodilators that activate its activity to cause MLC dephosphorylation and relaxation. The RhoA/ROCK pathway is activated by vasoconstrictors to inhibit myosin phosphatase activity. The mechanism by which RhoA and ROCK are localized to and interact with myosin light chain phosphatase (MLCP) is not well understood. We recently found a new member of the myosin phosphatase complex, myosin phosphatase-rho interacting protein, that directly binds to both RhoA and the myosin-binding subunit of myosin phosphatase in vitro, and targets myosin phosphatase to the actinomyosin contractile filament in smooth muscle cells. Because myosin phosphatase-rho interacting protein binds both RhoA and MLCP, we investigated whether myosin phosphatase-rho interacting protein was required for RhoA/ROCK-mediated myosin phosphatase regulation. Myosin phosphatase-rho interacting protein silencing prevented LPA-mediated myosin-binding subunit phosphorylation, and inhibition of myosin phosphatase activity. Myosin phosphatase-rho interacting protein did not regulate the activation of RhoA or ROCK in vascular smooth muscle cells. Silencing of M-RIP lead to loss of stress fiber-associated RhoA, suggesting that myosin phosphatase-rho interacting protein is a scaffold linking RhoA to regulate myosin phosphatase at the stress fiber.


Asunto(s)
Proteínas de Microfilamentos/fisiología , Contracción Muscular/fisiología , Miocitos del Músculo Liso/fisiología , Fosfatasa de Miosina de Cadena Ligera/fisiología , Quinasas Asociadas a rho/fisiología , Proteína de Unión al GTP rhoA/fisiología , Animales , Línea Celular , Activación Enzimática , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Miocitos del Músculo Liso/enzimología , Cadenas Ligeras de Miosina/metabolismo , Fosforilación , Unión Proteica , Ratas , Fibras de Estrés/fisiología
13.
Am J Pathol ; 171(2): 693-701, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17556591

RESUMEN

Pericytes regulate microvascular development and maturation through the control of endothelial cell motility, proliferation, and differentiation. The Rho GTPases have recently been described as key regulators of pericyte shape and contractile phenotype by signaling through the actin cytoskeleton in an isoactin-specific manner. In this report, we reveal that Rho GTPase-dependent signal transduction not only influences pericyte shape and contractile potential but also modulates capillary endothelial proliferative status and pericyte-endothelial interactions in vitro. We provide evidence that overexpression of mutant Rho GTPases, but not other Ras-related small GTPases, significantly alters pericyte shape, contractility, and endothelial growth state in microvascular cell co-cultures. In particular, we describe the use of a silicon substrate deformation assay to demonstrate that pericyte contractility is Rho GTP- and Rho kinase-dependent; further, we describe a novel in vitro system for examining pericyte-mediated endothelial growth arrest and show that control pericytes are capable of growth-arresting capillary endothelial cells in a cell contact-dependent manner, whereas pericytes overexpressing dominant-active and -negative Rho GTPase are comparably incompetent. These data strongly suggest that signaling through the pericyte Rho GTPase pathway may provide critical cues to the processes of microvascular stabilization, maturation, and contractility during development and disease.


Asunto(s)
Proliferación Celular , Forma de la Célula/fisiología , Endotelio Vascular/enzimología , Pericitos/enzimología , Proteínas de Unión al GTP rho/metabolismo , Amidas/farmacología , Animales , Animales Recién Nacidos , Capilares/citología , Capilares/fisiología , Bovinos , Forma de la Célula/efectos de los fármacos , Forma de la Célula/genética , Células Cultivadas , Técnicas de Cocultivo , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Microscopía de Contraste de Fase , Mutación , Pericitos/citología , Pericitos/fisiología , Piridinas/farmacología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/fisiología , Transfección , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Proteínas de Unión al GTP rho/genética
14.
J Biol Chem ; 280(52): 42543-51, 2005 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-16257966

RESUMEN

Vascular smooth muscle cell contraction and relaxation are directly related to the phosphorylation state of the regulatory myosin light chain. Myosin light chains are dephosphorylated by myosin phosphatase, leading to vascular smooth muscle relaxation. Myosin phosphatase is localized not only at actin-myosin stress fibers where it dephosphorylates myosin light chains, but also in the cytoplasm and at the cell membrane. The mechanisms by which myosin phosphatase is targeted to these loci are incompletely understood. We recently identified myosin phosphatase-Rho interacting protein as a member of the myosin phosphatase complex that directly binds both the myosin binding subunit of myosin phosphatase and RhoA and is localized to actin-myosin stress fibers. We hypothesized that myosin phosphatase-Rho interacting protein targets myosin phosphatase to the contractile apparatus to dephosphorylate myosin light chains. We used RNA interference to silence the expression of myosin phosphatase-Rho interacting protein in human vascular smooth muscle cells. Myosin phosphatase-Rho interacting protein silencing reduced the localization of the myosin binding subunit to stress fibers. This reduction in stress fiber myosin phosphatase-Rho interacting protein and myosin binding subunit increased basal and lysophosphatidic acid-stimulated myosin light chain phosphorylation. Neither cellular myosin phosphatase, myosin light chain kinase, nor RhoA activities were changed by myosin phosphatase-Rho interacting protein silencing. Furthermore, myosin phosphatase-Rho interacting protein silencing resulted in marked phenotypic changes in vascular smooth muscle cells, including increased numbers of stress fibers, increased cell area, and reduced stress fiber inhibition in response to a Rho-kinase inhibitor. These data support the importance of myosin phosphatase-Rho interacting protein-dependent targeting of myosin phosphatase to stress fibers for regulating myosin light chain phosphorylation state and morphology in human vascular smooth muscle cells.


Asunto(s)
Actinas/química , Proteínas Adaptadoras Transductoras de Señales/fisiología , Músculo Liso Vascular/citología , Cadenas Ligeras de Miosina/química , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Aorta/patología , Línea Celular , Células Cultivadas , Endotelio Vascular/patología , Inhibidores Enzimáticos/farmacología , Silenciador del Gen , Humanos , Microscopía Fluorescente , Modelos Genéticos , Contracción Muscular , Miocitos del Músculo Liso/citología , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Fenotipo , Fosforilación , Unión Proteica , Interferencia de ARN , Transfección , Proteína de Unión al GTP rhoA/metabolismo
15.
Proc Natl Acad Sci U S A ; 101(49): 17126-31, 2004 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-15569929

RESUMEN

Steroid hormone receptors (SHRs) are ligand-activated transcription factors that regulate gene expression. SHRs also mediate rapid, nongenomic cellular activation by steroids. In vascular endothelial cells, the SHR for estrogen, estrogen receptor (ER) alpha, is targeted by unknown mechanisms to a functional signaling module in membrane caveolae that enables estrogen to rapidly activate the mitogen-activated protein kinase and phosphatidylinositol 3-Akt kinase pathways, and endothelial NO synthase (eNOS). Here we identify the 110-kDa caveolin-binding protein striatin as the molecular anchor that localizes ERalpha to the membrane and organizes the ERalpha-eNOS membrane signaling complex. Striatin directly binds to amino acids 183-253 of ERalpha, targets ERalpha to the cell membrane, and serves as a scaffold for the formation of an ERalpha-Galphai complex. Disruption of complex formation between ERalpha and striatin blocks estrogen-induced rapid activation mitogen-activated protein kinase, Akt kinase, and eNOS, but has no effect on ER-dependent regulation of an estrogen response element-driven reporter plasmid. These findings identify striatin as a molecular scaffold required for rapid, nongenomic estrogen-mediated activation of downstream signaling pathways. Furthermore, by demonstrating independent regulation of nongenomic vs. genomic ER-dependent signaling, these findings provide conceptual support for the potential development of "pathway-specific" selective ER modulators.


Asunto(s)
Proteínas de Unión a Calmodulina/metabolismo , Receptor alfa de Estrógeno/fisiología , Sustancias Macromoleculares/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Óxido Nítrico Sintasa/metabolismo , Transducción de Señal , Proteínas de Unión a Calmodulina/fisiología , Caveolas/metabolismo , Línea Celular , Endotelio Vascular/citología , Activación Enzimática , Receptor alfa de Estrógeno/metabolismo , Estrógenos/farmacología , Humanos , Proteínas de la Membrana/fisiología , Proteínas del Tejido Nervioso/fisiología , Óxido Nítrico Sintasa de Tipo III , Unión Proteica
16.
J Biol Chem ; 279(40): 42055-61, 2004 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-15292222

RESUMEN

Excitation-contraction coupling in smooth muscle involves activation of myosin light chain (MLC) phosphorylation, which increases activity of the myosin actin-activated ATPase, resulting in contraction. Phosphorylation of MLC phosphatase (SMPP-1M) by Rho-associated kinase or endogenous SMPP-1M-associated kinase inhibits SMPP-1M, enhancing MLC phosphorylation and contraction. However, the precise identity of SMPP-1M-associated kinase remains unclear. Biochemical evidence strongly supports the idea that SMPP-1M-associated kinase is related to the human serine/threonine leucine zipper-interacting protein kinase (hZIPK), which is important in cell apoptosis, and the SMPP-1M-associated kinase has therefore been called ZIP-like kinase (MacDonald, J. A., Borman, M. A., Murani, A., Somlyo, A. V., Hartshorne, D. J., and Haystead, T. A. J. (2001) Proc. Natl. Acad. Sci. U. S. A. 98, 2419-2424). Whether the vascular smooth muscle SMPP-1M-associated kinase is a truncated version of hZIPK, native hZIPK, or a unique homologue of hZIPK is unclear. Here we show that only native hZIPK mRNA and protein are detectable in human vascular smooth muscle cells (VSMCs). High stringency screening of a human aortic cDNA library for the SMPP-1M-associated kinase identified 18 positive clones, all of which proved to be clones of hZIPK. PCR-based studies of VSMC RNA revealed native hZIPK transcripts but no evidence for splice variants of hZIPK or a ZIP-like kinase. Northern blotting studies of multiple vascular and non-vascular tissue RNAs, including human bladder RNA, showed only 2.3 kb of mRNA predicted for full-length hZIPK. Immunoblotting showed native full-length 52-kDa hZIPK expression in VSMCs. Full-length and N-terminal hZIPK bound the C-terminal domain (amino acids 681-847) of the myosin binding subunit (MBS) of SMPP-1M. hZIPK immunoprecipitated with the MBS of SMPP-1M and dominant negative RhoA inhibited the hZIPK-MBS interaction. These data identify hZIPK as the unique SMPP-1-associated kinase expressed in human vesicular smooth muscle and support a role for Rho in promoting the hZIPK-MBS interaction.


Asunto(s)
Músculo Liso Vascular/enzimología , Proteínas Serina-Treonina Quinasas/aislamiento & purificación , Secuencia de Aminoácidos , Proteínas Reguladoras de la Apoptosis , Proteínas Quinasas Dependientes de Calcio-Calmodulina , Línea Celular , Células Cultivadas , Proteínas Quinasas Asociadas a Muerte Celular , Humanos , Leucina Zippers , Músculo Liso Vascular/citología , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas/análisis , Proteínas Serina-Treonina Quinasas/metabolismo , Subunidades de Proteína , ARN Mensajero/análisis , Transfección , Proteína de Unión al GTP rhoA/fisiología
17.
J Biol Chem ; 279(23): 24420-6, 2004 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-15051728

RESUMEN

Cyclic GMP-dependent protein kinase I (PKGI) mediates vascular relaxation by nitric oxide and related nitrovasodilators and inhibits vascular smooth muscle cell (VSMC) migration. To identify VSMC proteins that interact with PKGI, the N-terminal protein interaction domain of PKGIalpha was used to screen a yeast two-hybrid human aortic cDNA library. The formin homology (FH) domain-containing protein, FHOD1, was found to interact with PKGIalpha in this screen. FH domain-containing proteins bind Rho-family GTPases and regulate actin cytoskeletal dynamics, cell migration, and gene expression. Antisera to FHOD1 were raised and used to characterize FHOD1 expression and distribution in vascular cells. FHOD1 is highly expressed in human coronary artery, aortic smooth muscle cells, and in human arterial and venous endothelial cells. In glutathione S-transferase pull-down experiments, the FHOD1 C terminus (amino acids 964-1165) binds full-length PKGI. Both in vitro and intact cell studies demonstrate that the interaction between FHOD1 and PKGI is decreased 3- to 5-fold in the presence of the PKG activator, 8Br-cGMP. Immunofluorescence studies of human VSMC show that FHOD1 is cytoplasmic and is concentrated in the perinuclear region. PKGI also directly phosphorylates FHOD1, and studies with wild-type and mutant FHOD1-derived peptides identify Ser-1131 in the FHOD1 C terminus as the unique PKGI phosphorylation site in FHOD1. These studies demonstrate that FHOD1 is a PKGI-interacting protein and substrate in VSMCs and show that cyclic GMP negatively regulates the FHOD1-PKGI interaction. Based on the known functions of FHOD1, the data are consistent with a role for FHOD1 in cyclic GMP-dependent inhibition of VSMC stress fiber formation and/or migration.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Proteínas Fetales/fisiología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Proteínas Nucleares/fisiología , Actinas/química , Animales , Western Blotting , Células COS , Movimiento Celular , Células Cultivadas , Centrifugación , Clonación Molecular , GMP Cíclico/metabolismo , Proteína Quinasa Dependiente de GMP Cíclico Tipo I , Proteínas Quinasas Dependientes de GMP Cíclico/química , Citoplasma/metabolismo , Citoesqueleto/metabolismo , ADN Complementario/metabolismo , Endotelio Vascular/metabolismo , Proteínas Fetales/química , Forminas , Expresión Génica , Biblioteca de Genes , Glutatión Transferasa/metabolismo , Humanos , Microscopía Confocal , Microscopía Fluorescente , Proteínas Nucleares/química , Pruebas de Precipitina , Unión Proteica , Estructura Terciaria de Proteína , Serina/química , Transfección , Técnicas del Sistema de Dos Híbridos
18.
J Biol Chem ; 278(51): 51484-93, 2003 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-14506264

RESUMEN

Regulation of vascular smooth muscle cell contractile state is critical for the maintenance of blood vessel tone. Abnormal vascular smooth muscle cell contractility plays an important role in the pathogenesis of hypertension, blood vessel spasm, and atherosclerosis. Myosin phosphatase, the key enzyme controlling myosin light chain dephosphorylation, regulates smooth muscle cell contraction. Vasoconstrictor and vasodilator pathways inhibit and activate myosin phosphatase, respectively. G-protein-coupled receptor agonists can inhibit myosin phosphatase and cause smooth muscle cell contraction by activating RhoA/Rho kinase, whereas NO/cGMP can activate myosin phosphatase and cause smooth muscle cell relaxation by activation of cGMP-dependent protein kinase. We have used yeast two-hybrid screening to identify a 116-kDa human protein that interacts with both myosin phosphatase and RhoA. This myosin phosphatase-RhoA interacting protein, or M-RIP, is highly homologous to murine p116RIP3, is expressed in vascular smooth muscle, and is localized to actin myofilaments. M-RIP binds directly to the myosin binding subunit of myosin phosphatase in vivo in vascular smooth muscle cells by an interaction between coiled-coil and leucine zipper domains in the two proteins. An adjacent domain of M-RIP directly binds RhoA in a nucleotide-independent manner. M-RIP copurifies with RhoA and Rho kinase, colocalizes on actin stress fibers with RhoA and MBS, and is associated with Rho kinase activity in vascular smooth muscle cells. M-RIP can assemble a complex containing both RhoA and MBS, suggesting that M-RIP may play a role in myosin phosphatase regulation by RhoA.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Citoesqueleto de Actina/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Células Cultivadas , Proteínas de Unión al GTP , Proteínas Activadoras de GTPasa/química , Humanos , Contracción Muscular , Músculo Liso Vascular/citología , Fosfatasa de Miosina de Cadena Ligera/química , Unión Proteica , Estructura Terciaria de Proteína , Subunidades de Proteína/metabolismo , Alineación de Secuencia , Técnicas del Sistema de Dos Híbridos
19.
Cell Signal ; 15(10): 937-44, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12873707

RESUMEN

Nitric oxide (NO) and nitrovasodilators induce vascular smooth muscle cell relaxation in part by cGMP-dependent protein kinase (cGK)-mediated activation of myosin phosphatase, which dephosphorylates myosin light chains. We recently found that cGMP-dependent protein kinase 1alpha binds directly to the myosin-binding subunit (MBS) of myosin phosphatase via the leucine/isoleucine zipper of cGK. We have now studied the role of the leucine zipper domain of MBS in dimerization with cGK and the leucine/isoleucine zipper and leucine zipper domains of both proteins in homodimerization. Mutagenesis of the MBS leucine zipper domain disrupts cGKIalpha-MBS dimerization. Mutagenesis of the MBS leucine zipper eliminates MBS homodimerization, while similar disruption of the cGKIalpha leucine/isoleucine zipper does not prevent formation of cGK dimers. The MBS leucine zipper domain is phosphorylated by cGK, but this does not have any apparent effect on heterodimer formation between the two proteins. MBS LZ mutants that are unable to bind cGK were poor substrates for cGK. These data support the theory that the MBS leucine zipper domain is necessary and sufficient to mediate both MBS homodimerization and binding of the protein to cGK. In contrast, the leucine/isoleucine zipper of cGK is required for binding to MBS, but not for cGK homodimerization. These data support that the MBS and cGK leucine zipper domains mediate the interaction between these two proteins. The contribution of these domains to both homodimerization and their specific interaction with each other suggest that additional regulatory mechanisms involving these domains may exist.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/química , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Miosinas/metabolismo , Secuencia de Aminoácidos , Animales , Proteína Quinasa Dependiente de GMP Cíclico Tipo I , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Dimerización , Leucina Zippers/fisiología , Datos de Secuencia Molecular , Miosinas/genética , Unión Proteica , Proteínas Recombinantes de Fusión/metabolismo
20.
J Biol Chem ; 278(7): 4639-45, 2003 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-12466266

RESUMEN

Estrogen receptor alpha (ERalpha) mediates the effects of estrogen by altering gene expression following hormone binding. It has recently been shown that kinase-mediated phosphorylation of ERalpha also transcriptionally activates the receptor in the absence of estrogen. We now report that ERalpha-dependent gene expression also is regulated by protein phosphatase 2A (PP2A). ERalpha co-immunoprecipitates with enzymatically active PP2A. ERalpha binds directly to the catalytic subunit of PP2A, which dephosphorylates serine 118 of the receptor. Amino acids 176-182 in the A/B domain of ERalpha are required for the interaction between PP2A and the receptor. Phosphatase inhibition disrupts the ERalpha-PP2A complex and induces formation of an ERalpha-activated mitogen-activated protein kinase complex, phosphorylation of ERalpha on serine 118, and transcriptional activation. These findings demonstrate that estrogen receptors exist in complexes with phosphatases as well as kinases. We propose a new model of ligand-independent activation of estrogen receptors in which the level of phosphorylation of ERalpha, and hence its transcriptional activation, is determined by the net effect of these counterregulatory pathways.


Asunto(s)
Fosfoproteínas Fosfatasas/metabolismo , Receptores de Estrógenos , Activación Transcripcional , Animales , Bovinos , Células Cultivadas , Receptor alfa de Estrógeno , Estrógenos/metabolismo , Humanos , Ligandos , Fosforilación , Proteína Fosfatasa 2 , Ratas , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA