Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Anticancer Agents Med Chem ; 21(9): 1092-1098, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32679023

RESUMEN

BACKGROUND: The Myosin Phosphatase (MP) holoenzyme is composed of a Protein Phosphatase type 1 (PP1) catalytic subunit and a regulatory subunit termed Myosin Phosphatase Target subunit 1 (MYPT1). Besides dephosphorylation of myosin, MP has been implicated in the control of cell proliferation via dephosphorylation and activation of the tumor suppressor gene products, retinoblastoma protein (pRb) and merlin. Inhibition of MP was shown to attenuate the drug-induced cell death of leukemic cells by chemotherapeutic agents, while activation of MP might have a sensitizing effect. OBJECTIVE: Recently, Epigallocatechin-Gallate (EGCG), a major component of green tea, was shown to activate MP by inducing the dephosphorylation of MYPT1 at phospho-Thr696 (MYPT1pT696), which might confer enhanced chemosensitivity to cancer cells. METHODS: THP-1 leukemic cells were treated with EGCG and Daunorubicin (DNR) and cell viability was analyzed. Phosphorylation of tumor suppressor proteins was detected by Western blotting. RESULTS: EGCG or DNR (at sub-lethal doses) alone had moderate effects on cell viability, while the combined treatment caused a significant decrease in the number of viable cells by enhancing apoptosis and decreasing proliferation. EGCG plus DNR decreased the phosphorylation level of MYPT1pT696, which was accompanied by prominent dephosphorylation of pRb. In addition, significant dephosphorylation of merlin was observed when EGCG and DNR were applied together. CONCLUSION: Our results suggest that EGCG-induced activation of MP might have a regulatory function in mediating the chemosensitivity of leukemic cells via dephosphorylation of tumor suppressor proteins.


Asunto(s)
Antineoplásicos/farmacología , Catequina/análogos & derivados , Daunorrubicina/farmacología , Fosfatasa de Miosina de Cadena Ligera/antagonistas & inhibidores , Antineoplásicos/síntesis química , Antineoplásicos/química , Catequina/farmacología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Daunorrubicina/síntesis química , Daunorrubicina/química , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Estructura Molecular , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Relación Estructura-Actividad , Células THP-1 , Células Tumorales Cultivadas
2.
J Biol Chem ; 294(36): 13280-13291, 2019 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-31315927

RESUMEN

Transforming growth factor-ß membrane associated protein (TIMAP) is an endothelial cell (EC)-predominant PP1 regulatory subunit and a member of the myosin phosphatase target (MYPT) protein family. The MYPTs preferentially bind the catalytic protein phosphatase 1 subunit PP1cß, forming myosin phosphatase holoenzymes. We investigated whether TIMAP/PP1cß could also function as a myosin phosphatase. Endogenous PP1cß, myosin light chain 2 (MLC2), and myosin IIA heavy chain coimmunoprecipitated from EC lysates with endogenous TIMAP, and endogenous MLC2 colocalized with TIMAP in EC projections. Purified recombinant GST-TIMAP interacted directly with purified recombinant His-MLC2. However, TIMAP overexpression in EC enhanced MLC2 phosphorylation, an effect not observed with a TIMAP mutant that does not bind PP1cß. Conversely, MLC2 phosphorylation was reduced in lung lysates from TIMAP-deficient mice and upon silencing of endogenous TIMAP expression in ECs. Ectopically expressed TIMAP slowed the rate of MLC2 dephosphorylation, an effect requiring TIMAP-PP1cß interaction. The association of MYPT1 with PP1cß was profoundly reduced in the presence of excess TIMAP, leading to proteasomal MYPT1 degradation. In the absence of TIMAP, MYPT1-associated PP1cß readily bound immobilized microcystin-LR, an active-site inhibitor of PP1c. By contrast, TIMAP-associated PP1cß did not interact with microcystin-LR, indicating that the active site of PP1cß is blocked when it is bound to TIMAP. Thus, TIMAP inhibits myosin phosphatase activity in ECs by competing with MYPT1 for PP1cß and blocking the PP1cß active site.


Asunto(s)
Proteínas de la Membrana/metabolismo , Fosfatasa de Miosina de Cadena Ligera/antagonistas & inhibidores , Proteína Fosfatasa 1/metabolismo , Animales , Biocatálisis , Línea Celular , Células Endoteliales/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Fosfatasa de Miosina de Cadena Ligera/metabolismo
3.
Auris Nasus Larynx ; 46(5): 790-796, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30739815

RESUMEN

Objective The endocytosis of cationized feritin (CF) via a clathrin-mediated pathway is regulated by a signaling network. Marginal cells showed the active endocytosis of CF via a clathrin-mediated pathway. The internalization of receptors through this clathrin-mediated pathway is an important regulatory event in signal transduction. Numerous kinases are involved in endocytosis, and each endocytic route is subjected to high-order regulation by cellular signaling mechanisms. In this study, we investigated whether ROCK and MLCK signaling cascades and G-proteins regulate the endocytosis of CF in marginal cells of the stria vascularis. Methods CF was infused into the cochlear duct with pertussis toxin (PTX),Clostridium botulinum C3 toxin (BTX), guanosine(g-thio)-triphosphate (GTP-γS), ML-7, Y-27632. Endocytic activity was measured at 30 min after the start of infusion under an electron microscope. Results In marginal cells, CF was internalized via a clathrin-mediated pathway that depends on F-actin and microtubules (MT). Its processes were controlled by myosin light chain kinase (MLCK) and Rho-associated kinase (ROCK), but not affected by G-protein-coupled receptor (GPCR) or the RhoA signaling cascade. Conclusion Our previous study showed that the main endocytotic pathway of microperoxidase (MPO) did not depend on the Rho/ROCK molecular switch or actin/myosin motor system, but was mainly regulated by the RhoA signaling cascade. The present study results indicate that these signaling cascades regulating CF internalization completely differ from the cascades for MPO internalization.


Asunto(s)
Vesículas Cubiertas por Clatrina/metabolismo , Endocitosis/fisiología , Ferritinas/metabolismo , Proteínas de Unión al GTP/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Estría Vascular/metabolismo , Quinasas Asociadas a rho/metabolismo , ADP Ribosa Transferasas/farmacología , Amidas/farmacología , Animales , Azepinas/farmacología , Toxinas Botulínicas/farmacología , Conducto Coclear , Endocitosis/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Proteínas de Unión al GTP/antagonistas & inhibidores , Cobayas , Microscopía Electrónica , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Fosfatasa de Miosina de Cadena Ligera/antagonistas & inhibidores , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Naftalenos/farmacología , Toxina del Pertussis/farmacología , Piridinas/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Estría Vascular/efectos de los fármacos , Quinasas Asociadas a rho/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/metabolismo
4.
Development ; 145(8)2018 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-29691225

RESUMEN

Epithelial folding shapes embryos and tissues during development. Here, we investigate the coupling between epithelial folding and actomyosin-enriched compartmental boundaries. The mechanistic relationship between the two is unclear, because actomyosin-enriched boundaries are not necessarily associated with folds. Also, some cases of epithelial folding occur independently of actomyosin contractility. We investigated the shallow folds called parasegment grooves that form at boundaries between anterior and posterior compartments in the early Drosophila embryo. We demonstrate that formation of these folds requires the presence of an actomyosin enrichment along the boundary cell-cell contacts. These enrichments, which require Wingless signalling, increase interfacial tension not only at the level of the adherens junctions but also along the lateral surfaces. We find that epithelial folding is normally under inhibitory control because different genetic manipulations, including depletion of the Myosin II phosphatase Flapwing, increase the depth of folds at boundaries. Fold depth correlates with the levels of Bazooka (Baz), the Par-3 homologue, along the boundary cell-cell contacts. Moreover, Wingless and Hedgehog signalling have opposite effects on fold depth at the boundary that correlate with changes in Baz planar polarity.


Asunto(s)
Actomiosina/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriología , Drosophila melanogaster/metabolismo , Proteína Wnt1/metabolismo , Uniones Adherentes/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas Bacterianas/genética , Tipificación del Cuerpo , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Epitelio/embriología , Técnicas de Silenciamiento del Gen , Genes de Insecto , Proteínas Fluorescentes Verdes/genética , Proteínas Hedgehog/antagonistas & inhibidores , Proteínas Hedgehog/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Luminiscentes/genética , Mutación , Miosina Tipo II/metabolismo , Fosfatasa de Miosina de Cadena Ligera/antagonistas & inhibidores , Fosfatasa de Miosina de Cadena Ligera/genética , Transducción de Señal , Proteína Wnt1/genética
5.
Int J Mol Med ; 41(1): 430-438, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29115372

RESUMEN

The aim of the present study was to investigate the role of Rho kinase (also known as ROCK) inhibitor in 2,4,6-trinitrobenzene sulfonic acid induced mouse colitis; and to elucidate the underlying mechanism of ROCK1/ROCK2 inhibition in enhancing intestinal epithelial barrier (IEB) function. A specific inhibitor of ROCK, Y-27632, was used to examine the role of ROCK in mouse colitis models. ROCK1 and ROCK2 were silenced respectively using RNA interference in Caco-2 cells. The expression of tight junction proteins and the downstream molecules of ROCK were assessed. Y-27632 alleviated colonic inflammation and decreased intestinal permeability. ROCK-myosin light chain (MLC) and ROCK-NF-κB pathway were activated in colitis and inhibited by Y-27632. In vitro, ROCK1 RNAi primarily downregulated the phosphorylation of myosin phosphatase-targeting subunit-1 (MYPT-1) and MLC, while ROCK2 RNAi inhibited phosphorylation of nuclear factor-κB (NF-κB). In conclusion, the results suggested that the ROCK inhibitor alleviated colitis and IEB dysfunction. Inhibition of phospho-MYPT-1 and MLC by ROCK1 knockout or inhibition of NF-κB phosphorylation by ROCK2 knockout may be the underlying mechanisms.


Asunto(s)
Colitis/tratamiento farmacológico , Mucosa Intestinal/metabolismo , Quinasas Asociadas a rho/genética , Amidas/administración & dosificación , Animales , Células CACO-2 , Colitis/inducido químicamente , Colitis/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Mucosa Intestinal/efectos de los fármacos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Fosfatasa de Miosina de Cadena Ligera/antagonistas & inhibidores , Fosfatasa de Miosina de Cadena Ligera/genética , Piridinas/administración & dosificación , Ácido Trinitrobencenosulfónico/toxicidad , Quinasas Asociadas a rho/antagonistas & inhibidores
6.
J Cereb Blood Flow Metab ; 37(3): 1014-1029, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27193035

RESUMEN

Aging causes major alterations of all components of the neurovascular unit and compromises brain blood supply. Here, we tested how aging affects vascular reactivity in basilar arteries from young (<10 weeks; y-BA), old (>22 months; o-BA) and old (>22 months) heterozygous MYPT1-T-696A/+ knock-in mice. In isometrically mounted o-BA, media thickness was increased by ∼10% while the passive length tension relations were not altered. Endothelial denudation or pan-NOS inhibition (100 µmol/L L-NAME) increased the basal tone by 11% in y-BA and 23% in o-BA, while inhibition of nNOS (1 µmol/L L-NPA) induced ∼10% increase in both ages. eNOS expression was ∼2-fold higher in o-BA. In o-BA, U46619-induced force was augmented (pEC50 ∼6.9 vs. pEC50 ∼6.5) while responsiveness to DEA-NONOate, electrical field stimulation or nicotine was decreased. Basal phosphorylation of MLC20-S19 and MYPT1-T-853 was higher in o-BA and was reversed by apocynin. Furthermore, permeabilized o-BA showed enhanced Ca2+-sensitivity. Old T-696A/+ BA displayed a reduced phosphorylation of MYPT1-T696 and MLC20, a lower basal tone in response to L-NAME and a reduced eNOS expression. The results indicate that the vascular hypercontractility found in o-BA is mediated by inhibition of MLCP and is partially compensated by an upregulation of endothelial NO release.


Asunto(s)
Acetofenonas/farmacología , Envejecimiento , Arteria Basilar/fisiología , Músculo Liso Vascular/fisiología , Fosfatasa de Miosina de Cadena Ligera/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/metabolismo , Animales , Inhibidores Enzimáticos , Ratones , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Fosforilación , Subunidades de Proteína/metabolismo , Vasoconstricción
7.
Mol Neurobiol ; 54(1): 154-168, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-26732598

RESUMEN

The loss of endothelial cells (ECs) homeostasis is a trigger for cerebrovascular dysfunction that is a common event in several neurodegenerative disorders such as Alzheimer's disease (AD). The present work addressed the role of phosphatase 2A (PP2A) in cytoskeleton rearrangement, endoplasmic reticulum (ER) homeostasis, ER-mitochondria communication and mitochondrial dynamics in brain ECs. For this purpose, rat brain endothelial (RBE4) cells were exposed to okadaic acid, a well-known inhibitor of PP2A activity. An increase in the levels of tau phosphorylated on Ser396 and Thr181 residues was observed upon PP2A inhibition, concomitantly with the rearrangement of microtubules and actin cytoskeleton. Under these conditions, an increase in the levels of ER stress markers, namely GRP78, XBP1, p-eIF2αSer51, and ERO1α, was observed. Moreover, PP2A inhibition upregulated the Sigma-1 receptor, an ER chaperone located at the ER-mitochondria interface, and enhanced inter-organelle Ca2+ transfer, culminating in mitochondrial Ca2+ overload and activation of mitochondria-dependent apoptosis. The inhibition of PP2A activity also promoted an alteration of the structural and spatial mitochondria network due to upregulation of mitochondrial fission (Drp1 and Fis1) and fusion (Mfn1, Mfn2 and OPA1) proteins, suggesting detrimental changes in mitochondrial dynamics. In accordance with our in vitro observations, brain vessels from 3xTg-AD mice showed a significant decrease in PP2A protein levels accompanied by an increase in tau phosphorylated on Ser396 and GRP78 protein levels. Collectively, these results suggest that the loss of cerebrovascular homeostasis that occurs in AD might be a downstream event of the compromised activity and/or expression of PP2A, which is observed in the brain of individuals affected with this devastating neurodegenerative disorder.


Asunto(s)
Encéfalo/enzimología , Citoesqueleto/enzimología , Retículo Endoplásmico/enzimología , Células Endoteliales/enzimología , Mitocondrias/enzimología , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Animales , Encéfalo/citología , Encéfalo/efectos de los fármacos , Línea Celular , Citoesqueleto/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Retículo Endoplásmico/efectos de los fármacos , Chaperón BiP del Retículo Endoplásmico , Células Endoteliales/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Masculino , Ratones , Ratones Transgénicos , Mitocondrias/efectos de los fármacos , Fosfatasa de Miosina de Cadena Ligera/antagonistas & inhibidores , Ácido Ocadaico/farmacología , Ratas
8.
J Cereb Blood Flow Metab ; 37(1): 227-240, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-26721393

RESUMEN

The myogenic response of cerebral resistance arterial smooth muscle to intraluminal pressure elevation is a key physiological mechanism regulating blood flow to the brain. Rho-associated kinase plays a critical role in the myogenic response by activating Ca2+ sensitization mechanisms: (i) Rho-associated kinase inhibits myosin light chain phosphatase by phosphorylating its targeting subunit myosin phosphatase targeting subunit 1 (at T855), augmenting 20 kDa myosin regulatory light chain (LC20) phosphorylation and force generation; and (ii) Rho-associated kinase stimulates cytoskeletal actin polymerization, enhancing force transmission to the cell membrane. Here, we tested the hypothesis that abnormal Rho-associated kinase-mediated myosin light chain phosphatase regulation underlies the dysfunctional cerebral myogenic response of the Goto-Kakizaki rat model of type 2 diabetes. Basal levels of myogenic tone, LC20, and MYPT1-T855 phosphorylation were elevated and G-actin content was reduced in arteries of pre-diabetic 8-10 weeks Goto-Kakizaki rats with normal serum insulin and glucose levels. Pressure-dependent myogenic constriction, LC20, and myosin phosphatase targeting subunit 1 phosphorylation and actin polymerization were suppressed in both pre-diabetic Goto-Kakizaki and diabetic (18-20 weeks) Goto-Kakizaki rats, whereas RhoA, ROK2, and MYPT1 expression were unaffected. We conclude that abnormal Rho-associated kinase-mediated Ca2+ sensitization contributes to the dysfunctional cerebral myogenic response in the Goto-Kakizaki model of type 2 diabetes.


Asunto(s)
Actinas/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Fosfatasa de Miosina de Cadena Ligera/antagonistas & inhibidores , Vasoconstricción , Animales , Calcio/metabolismo , Arterias Cerebrales/fisiopatología , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Fosforilación , Polimerizacion , Ratas , Ratas Endogámicas , Quinasas Asociadas a rho
9.
J Pharmacol Exp Ther ; 356(1): 53-63, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26493746

RESUMEN

Endogenous hydrogen sulfide (H2S) is involved in the regulation of vascular tone. We hypothesized that the lowering of calcium and opening of potassium (K) channels as well as calcium-independent mechanisms are involved in H2S-induced relaxation in rat mesenteric small arteries. Amperometric recordings revealed that free [H2S] after addition to closed tubes of sodium hydrosulfide (NaHS), Na2S, and GYY4137 [P-(4-methoxyphenyl)-P-4-morpholinyl-phosphinodithioic acid] were, respectively, 14%, 17%, and 1% of added amount. The compounds caused equipotent relaxations in isometric myographs, but based on the measured free [H2S], GYY4137 caused more relaxation in relation to released free H2S than NaHS and Na2S in rat mesenteric small arteries. Simultaneous measurements of [H2S] and tension showed that 15 µM of free H2S caused 61% relaxation in superior mesenteric arteries. Simultaneous measurements of smooth muscle calcium and tension revealed that NaHS lowered calcium and caused relaxation of NE-contracted arteries, while high extracellular potassium reduced NaHS relaxation without corresponding calcium changes. In NE-contracted arteries, NaHS (1 mM) lowered the phosphorylation of myosin light chain, while phosphorylation of myosin phosphatase target subunit 1 remained unchanged. Protein kinase A and G, inhibitors of guanylate cyclase, failed to reduce NaHS relaxation, whereas blockers of voltage-gated KV7 channels inhibited NaHS relaxation, and blockers of mitochondrial complex I and III abolished NaHS relaxation. Our findings suggest that low micromolar concentrations of free H2S open K channels followed by lowering of smooth muscle calcium, and by another mechanism involving mitochondrial complex I and III leads to uncoupling of force, and hence vasodilation.


Asunto(s)
Calcio/metabolismo , Sulfuro de Hidrógeno/farmacología , Arterias Mesentéricas/efectos de los fármacos , Relajación Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Canales de Potasio/efectos de los fármacos , Animales , Complejo I de Transporte de Electrón/efectos de los fármacos , Complejo III de Transporte de Electrones/antagonistas & inhibidores , Sulfuro de Hidrógeno/metabolismo , Técnicas In Vitro , Canales de Potasio KCNQ/efectos de los fármacos , Arterias Mesentéricas/metabolismo , Músculo Liso Vascular/metabolismo , Cadenas Ligeras de Miosina/efectos de los fármacos , Cadenas Ligeras de Miosina/metabolismo , Fosfatasa de Miosina de Cadena Ligera/antagonistas & inhibidores , Fosforilación , Bloqueadores de los Canales de Potasio/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Ratas Wistar , Vasodilatación/efectos de los fármacos
10.
Basic Clin Pharmacol Toxicol ; 119 Suppl 3: 86-95, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26708952

RESUMEN

Diabetes is associated with erectile dysfunction and with hypercontractility in erectile tissue and this is in part ascribed to increased formation of thromboxane. Rho kinase (ROCK) is a key regulator of calcium sensitization and contraction in vascular smooth muscle. This study investigated the role of calcium and ROCK in contraction evoked by activation of the thromboxane receptors. Rat intracavernous penile arteries were mounted for isometric tension and intracellular calcium ([Ca2+ ]i ) recording and corpus cavernosum for measurements of MYPT1 phosphorylation. In penile arteries, U46619 by activation of thromboxane receptors concentration dependently increased calcium and contraction. U46619-induced calcium influx was blocked by nifedipine, a blocker of L-type calcium channels, and by 2-aminoethoxydiphenyl borate, a blocker of transient receptor potential (TRP) channels. Inhibitors of ROCK, Y27632 and glycyl-H1152P, concentration dependently reduced U46619-induced contraction, but only Y27632 reduced [Ca2+ ]i levels in the penile arteries activated with either high extracellular potassium or U46619. MYPT-Thr850 phosphorylation in corpus cavernous strips was increased in response to U46619 through activation of TP receptors and was found to be a direct result of phosphorylation by ROCK. Y27632 induced less relaxation in mesenteric arteries, H1152P induced equipotent relaxations, and a protein kinase C inhibitor, Ro-318220, failed to relax intracavernous penile arteries, but induced full relaxation in rat mesenteric arteries. Our findings suggest that U46619 contraction depends on Ca2+ influx through L-type and TRP channels, and ROCK-dependent mechanisms in penile arteries. Inhibition of the ROCK pathway is a potential approach for the treatment of erectile dysfunction associated with hypertension and diabetes.


Asunto(s)
Arterias/fisiología , Microcirculación , Músculo Liso Vascular/irrigación sanguínea , Pene/irrigación sanguínea , Receptores de Tromboxano A2 y Prostaglandina H2/agonistas , Tromboxano A2/metabolismo , Quinasas Asociadas a rho/metabolismo , Animales , Arterias/efectos de los fármacos , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/química , Canales de Calcio Tipo L/metabolismo , Señalización del Calcio/efectos de los fármacos , Técnicas In Vitro , Masculino , Microcirculación/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Fosfatasa de Miosina de Cadena Ligera/antagonistas & inhibidores , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Especificidad de Órganos , Pene/efectos de los fármacos , Pene/metabolismo , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ratas Wistar , Receptores de Tromboxano A2 y Prostaglandina H2/metabolismo , Canales de Potencial de Receptor Transitorio/antagonistas & inhibidores , Canales de Potencial de Receptor Transitorio/metabolismo , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/antagonistas & inhibidores , Vasoconstrictores/farmacología , Vasodilatadores/farmacología , Quinasas Asociadas a rho/antagonistas & inhibidores
11.
Biol Pharm Bull ; 38(11): 1809-16, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26521832

RESUMEN

Orthovanadate (OVA), a protein tyrosine phosphatase inhibitor, induces vasoconstriction in a Rho kinase-dependent manner. The aim of this study was to determine the mechanism underlying OVA-induced vasoconstriction of rat mesenteric arteries. OVA-induced constriction of mesenteric arterial rings treated with N(G)-nitro-L-arginine methyl ester (L-NAME, 0.1 mM), a nitric oxide synthase inhibitor, was significantly blocked by the Rho kinase inhibitor Y-27632 (R-(+)-trans-N-(4-pyridyl)-4-(1-aminoethyl)-cyclohexanecarboxamide, 10 µM), extracellular signal-regulated kinase 1 and 2 (Erk1/2) inhibitor FR180204 (5-(2-phenyl-pyrazolo[1,5-a]pyridin-3-yl)-1H-pyrazolo[3,4-c]pyridazin-3-ylamine, 10 µM), Erk1/2 kinase (MEK) inhibitor PD98059 (2'-amino-3'-methoxyflavone, 10 µM), epidermal growth factor receptor (EGFR) inhibitor AG1478 (4-(3-chloroanilino)-6,7-dimethoxyquinazoline, 10 µM), and Src inhibitor PP2 (4-amino-3-(4-chlorophenyl)-1-(t-butyl)-1H-pyrazolo[3,4-d]pyrimidine, 3 µM). However, the myosin light chain kinase inhibitor ML-7 (1-(5-iodonaphthalene-1-sulfonyl)-homopiperazine, 10 µM) did not affect OVA-induced constriction. Phosphorylation of myosin phosphatase target subunit 1 (MYPT1, an index of Rho kinase activity) was abrogated by inhibitors of Src, EGFR MEK, Erk1/2, and Rho kinase. OVA-stimulated Erk1/2 phosphorylation was blocked by inhibitors of EGFR, Src, MEK, and Erk1/2 but not affected by an inhibitor of Rho kinase. OVA-induced Src phosphorylation was abrogated by an Src inhibitor but not affected by inhibitors of EGFR, MEK, Erk1/2, and Rho kinase. In addition, the metalloproteinase inhibitor TAPI-0 (N-(R)-[2-(hydroxyaminocarbonyl)methyl]-4-methylpentanoyl-L-naphthylalanyl-L-alanine amide, 10 µM) and an inhibitor of heparin/epidermal growth factor binding (CRM 197, 10 µg/mL) did not affect OVA-induced contraction of rat mesenteric arterial rings. These results suggest that OVA induces vasoconstriction in rat mesenteric arteries via Src, EGFR, MEK, and Erk1/2 activation, leading to the inactivation of myosin light chain phosphatase through phosphorylation of MYPT1.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Arterias Mesentéricas/efectos de los fármacos , Mesenterio/efectos de los fármacos , Fosfatasa de Miosina de Cadena Ligera/antagonistas & inhibidores , Vanadatos/farmacología , Vasoconstricción/efectos de los fármacos , Quinasas Asociadas a rho/metabolismo , Animales , Receptores ErbB/metabolismo , Masculino , Arterias Mesentéricas/metabolismo , Mesenterio/irrigación sanguínea , Mesenterio/metabolismo , Fosforilación , Piperazina , Piperazinas/farmacología , Proteína Fosfatasa 1/metabolismo , Ratas Wistar , Transducción de Señal , Vanadio/metabolismo , Familia-src Quinasas/metabolismo
12.
Circ Res ; 116(5): 895-908, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25722443

RESUMEN

Vascular stiffness is a mechanical property of the vessel wall that affects blood pressure, permeability, and inflammation. As a result, vascular stiffness is a key driver of (chronic) human disorders, including pulmonary arterial hypertension, kidney disease, and atherosclerosis. Responses of the endothelium to stiffening involve integration of mechanical cues from various sources, including the extracellular matrix, smooth muscle cells, and the forces that derive from shear stress of blood. This response in turn affects endothelial cell contractility, which is an important property that regulates endothelial stiffness, permeability, and leukocyte-vessel wall interactions. Moreover, endothelial stiffening reduces nitric oxide production, which promotes smooth muscle cell contraction and vasoconstriction. In fact, vessel wall stiffening, and microcirculatory endothelial dysfunction, precedes hypertension and thus underlies the development of vascular disease. Here, we review the cross talk among vessel wall stiffening, endothelial contractility, and vascular disease, which is controlled by Rho-driven actomyosin contractility and cellular mechanotransduction. In addition to discussing the various inputs and relevant molecular events in the endothelium, we address which actomyosin-regulated changes at cell adhesion complexes are genetically associated with human cardiovascular disease. Finally, we discuss recent findings that broaden therapeutic options for targeting this important mechanical signaling pathway in vascular pathogenesis.


Asunto(s)
Enfermedades Cardiovasculares/fisiopatología , Endotelio Vascular/fisiopatología , Mecanotransducción Celular/fisiología , Rigidez Vascular/fisiología , Quinasas Asociadas a rho/fisiología , Actomiosina/fisiología , Envejecimiento/fisiología , Animales , Calcinosis/patología , Calcinosis/fisiopatología , Enfermedades Cardiovasculares/enzimología , Adhesión Celular/fisiología , Permeabilidad de la Membrana Celular , Citoesqueleto/ultraestructura , Endotelio Vascular/ultraestructura , Hemorreología , Humanos , Inflamación , Integrinas/fisiología , Leucocitos/fisiología , Mecanotransducción Celular/efectos de los fármacos , Ratones , Ratones Noqueados , Microcirculación , Modelos Cardiovasculares , Fosfatasa de Miosina de Cadena Ligera/antagonistas & inhibidores , Fosfatasa de Miosina de Cadena Ligera/fisiología , FN-kappa B/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Análisis de la Onda del Pulso , Ratas , Migración Transendotelial y Transepitelial , Rigidez Vascular/efectos de los fármacos , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Proteínas de Unión al GTP rho/fisiología , Quinasas Asociadas a rho/antagonistas & inhibidores
13.
J Exp Med ; 212(2): 267-80, 2015 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-25601651

RESUMEN

Neutrophils respond to invading bacteria by adopting a polarized morphology, migrating in the correct direction, and engulfing the bacteria. How neutrophils establish and precisely orient this polarity toward pathogens remains unclear. Here we report that in resting neutrophils, the ERM (ezrin, radixin, and moesin) protein moesin in its active form (phosphorylated and membrane bound) prevented cell polarization by inhibiting the small GTPases Rac, Rho, and Cdc42. Attractant-induced activation of myosin phosphatase deactivated moesin at the prospective leading edge to break symmetry and establish polarity. Subsequent translocation of moesin to the trailing edge confined the formation of a prominent pseudopod directed toward pathogens and prevented secondary pseudopod formation in other directions. Therefore, both moesin-mediated inhibition and its localized deactivation by myosin phosphatase are essential for neutrophil polarization and effective neutrophil tracking of pathogens.


Asunto(s)
Quimiotaxis de Leucocito , Proteínas de Microfilamentos/metabolismo , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Animales , Línea Celular , Quimiotaxis de Leucocito/genética , Quimiotaxis de Leucocito/inmunología , Eliminación de Gen , Técnicas de Silenciamiento del Gen , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Masculino , Ratones , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Fosfatasa de Miosina de Cadena Ligera/antagonistas & inhibidores , Infiltración Neutrófila/genética , Infiltración Neutrófila/inmunología , Neutrófilos/microbiología , Fagocitosis/genética , Fagocitosis/inmunología , Fosforilación , Unión Proteica , Interferencia de ARN , Factores de Intercambio de Guanina Nucleótido Rho/genética , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Proteína de Unión al GTP cdc42/antagonistas & inhibidores , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rac/antagonistas & inhibidores , Proteínas de Unión al GTP rac/metabolismo , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Proteínas de Unión al GTP rho/metabolismo
14.
J Physiol ; 591(5): 1235-50, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23230233

RESUMEN

Abstract The myogenic response of resistance arteries to intravascular pressure elevation is a fundamental physiological mechanism of crucial importance for blood pressure regulation and organ-specific control of blood flow. The importance of Ca(2+) entry via voltage-gated Ca(2+) channels leading to phosphorylation of the 20 kDa myosin regulatory light chains (LC20) in the myogenic response is well established. Recent studies, however, have suggested a role for Ca(2+) sensitization via activation of the RhoA/Rho-associated kinase (ROK) pathway in the myogenic response. The possibility that enhanced actin polymerization is also involved in myogenic vasoconstriction has been suggested. Here, we have used pressurized resistance arteries from rat gracilis and cremaster skeletal muscles to assess the contribution to myogenic constriction of Ca(2+) sensitization due to: (1) phosphorylation of the myosin targeting subunit of myosin light chain phosphatase (MYPT1) by ROK; (2) phosphorylation of the 17 kDa protein kinase C (PKC)-potentiated protein phosphatase 1 inhibitor protein (CPI-17) by PKC; and (3) dynamic reorganization of the actin cytoskeleton evoked by ROK and PKC. Arterial diameter, MYPT1, CPI-17 and LC20 phosphorylation, and G-actin content were determined at varied intraluminal pressures ± H1152, GF109203X or latrunculin B to suppress ROK, PKC and actin polymerization, respectively. The myogenic response was associated with an increase in MYPT1 and LC20 phosphorylation that was blocked by H1152. No change in phospho-CPI-17 content was detected although the PKC inhibitor, GF109203X, suppressed myogenic constriction. Basal LC20 phosphorylation at 10 mmHg was high at ∼40%, increased to a maximal level of ∼55% at 80 mmHg, and exhibited no additional change on further pressurization to 120 and 140 mmHg. Myogenic constriction at 80 mmHg was associated with a decline in G-actin content by ∼65% that was blocked by inhibition of ROK or PKC. Taken together, our findings indicate that two mechanisms of Ca(2+) sensitization (ROK-mediated phosphorylation of MYPT1-T855 with augmentation of LC20 phosphorylation, and a ROK- and PKC-evoked increase in actin polymerization) contribute to force generation in the myogenic response of skeletal muscle arterioles.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Músculo Esquelético/irrigación sanguínea , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Resistencia Vascular , Vasoconstricción , Citoesqueleto de Actina/efectos de los fármacos , Animales , Presión Arterial , Arterias/enzimología , Señalización del Calcio , Masculino , Mecanotransducción Celular , Proteínas Musculares/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Fosfatasa de Miosina de Cadena Ligera/antagonistas & inhibidores , Fosfoproteínas/metabolismo , Fosforilación , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteína Fosfatasa 1/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Resistencia Vascular/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
15.
Am J Physiol Cell Physiol ; 303(1): C58-68, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22538237

RESUMEN

Cellular activity of the myosin light chain phosphatase (MLCP) determines agonist-induced force development of smooth muscle (SM). CPI-17 is an endogenous inhibitor protein for MLCP, responsible for mediating G-protein signaling into SM contraction. Fluctuations in CPI-17 expression occur in response to pathological stresses, altering excitation-contraction coupling in SM. Here, we determined the signaling pathways regulating CPI-17 expression in rat aorta tissues and the cell culture using a pharmacological approach. CPI-17 transcription was suppressed in response to the proliferative stimulus with platelet-derived growth factor (PDGF) through the ERK1/2 pathway, whereas it was elevated in response to inflammatory, stress-induced and excitatory stimuli with transforming growth factor-ß, IL-1ß, TNFα, sorbitol, and serotonin. CPI-17 transcription was repressed by inhibition of JNK, p38, PKC, and Rho-kinase (ROCK). The mouse and human CPI-17 gene promoters were governed by the proximal GC-boxes at the 5'-flanking region, where Sp1/Sp3 transcription factors bound. Sp1 binding to the region was more prominent in intact aorta tissues, compared with the SM cell culture, where the CPI-17 gene is repressed. The 173-bp proximal promoter activity was negatively and positively regulated through PDGF-induced ERK1/2 and sorbitol-induced p38/JNK pathways, respectively. By contrast, PKC and ROCK inhibitors failed to repress the 173-bp promoter activity, suggesting distal enhancer elements. CPI-17 transcription was insensitive to knockdown of myocardin/Kruppel-like factor 4 small interfering RNA or histone deacetylase inhibition. The reciprocal regulation of Sp1/Sp3-driven CPI-17 expression through multiple kinases may be responsible for the adaptation of MLCP signal and SM tone to environmental changes.


Asunto(s)
Proteínas Musculares/metabolismo , Músculo Liso Vascular/metabolismo , Fosfatasa de Miosina de Cadena Ligera/antagonistas & inhibidores , Fosfoproteínas/metabolismo , Animales , Aorta/metabolismo , Células Cultivadas , Humanos , Interleucina-1beta/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Contracción Muscular/fisiología , Músculo Liso Vascular/citología , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Proteínas Nucleares/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Regiones Promotoras Genéticas , Proteína Quinasa C/antagonistas & inhibidores , Interferencia de ARN , ARN Interferente Pequeño , Ratas , Ratas Sprague-Dawley , Serotonina/metabolismo , Transducción de Señal , Sorbitol/metabolismo , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción Sp3/metabolismo , Transactivadores/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Quinasas Asociadas a rho/antagonistas & inhibidores
16.
Cell Mol Life Sci ; 69(2): 247-66, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21947498

RESUMEN

Vascular smooth muscle tone is controlled by a balance between the cellular signaling pathways that mediate the generation of force (vasoconstriction) and release of force (vasodilation). The initiation of force is associated with increases in intracellular calcium concentrations, activation of myosin light-chain kinase, increases in the phosphorylation of the regulatory myosin light chains, and actin-myosin crossbridge cycling. There are, however, several signaling pathways modulating Ca(2+) mobilization and Ca(2+) sensitivity of the contractile machinery that secondarily regulate the contractile response of vascular smooth muscle to receptor agonists. Among these regulatory mechanisms involved in the physiological regulation of vascular tone are the cyclic nucleotides (cAMP and cGMP), which are considered the main messengers that mediate vasodilation under physiological conditions. At least four distinct mechanisms are currently thought to be involved in the vasodilator effect of cyclic nucleotides and their dependent protein kinases: (1) the decrease in cytosolic calcium concentration ([Ca(2+)]c), (2) the hyperpolarization of the smooth muscle cell membrane potential, (3) the reduction in the sensitivity of the contractile machinery by decreasing the [Ca(2+)]c sensitivity of myosin light-chain phosphorylation, and (4) the reduction in the sensitivity of the contractile machinery by uncoupling contraction from myosin light-chain phosphorylation. This review focuses on each of these mechanisms involved in cyclic nucleotide-dependent relaxation of vascular smooth muscle under physiological conditions.


Asunto(s)
Músculo Liso Vascular/efectos de los fármacos , Nucleótidos Cíclicos/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Animales , Calcio/metabolismo , Calcio/fisiología , ATPasas Transportadoras de Calcio/metabolismo , ATPasas Transportadoras de Calcio/fisiología , Humanos , Ratones , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología , Músculo Liso Vascular/fisiología , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/fisiología , Fosfatasa de Miosina de Cadena Ligera/antagonistas & inhibidores , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Fosfatasa de Miosina de Cadena Ligera/fisiología , Nucleótidos Cíclicos/metabolismo , Nucleótidos Cíclicos/fisiología , Canales de Potasio/agonistas , Canales de Potasio/metabolismo , Canales de Potasio/fisiología , Ratas , Retículo Sarcoplasmático/efectos de los fármacos , Retículo Sarcoplasmático/metabolismo , Retículo Sarcoplasmático/fisiología , Intercambiador de Sodio-Calcio/metabolismo , Intercambiador de Sodio-Calcio/fisiología , Vasodilatación/fisiología , Vasodilatadores/metabolismo
17.
Pharmacology ; 88(1-2): 65-71, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21846997

RESUMEN

Peroxisome proliferator-activated receptor α (PPAR-α) is a ligand-activated transcription factor that exerts strong effects on metabolic pathways. Our aim was to elucidate the effect of clofibrate, a PPAR-α agonist, on the longitudinal muscle of the mouse distal colon. We initially found that clofibrate induced a relaxation response in this muscle. Notably, the PPAR-α antagonists GW9662 and T0070907 did not attenuate this clofibrate-induced relaxation. The structurally related PPAR-α agonists fenofibrate and bezafibrate induced relaxation in the distal colon as effectively as clofibrate. In contrast, wy-14643, which activates PPAR-α more selectively than clofibrate, had no effect. Furthermore, clofibrate-induced relaxation was not affected by N-nitro-L-arginine, an NO synthase inhibitor, 1H-[1,2,4]-oxadiazolo-[4,3- a]quinoxaline-1-one, a soluble guanylate cyclase inhibitor, or H89, a protein kinase A inhibitor. Tetrodotoxin, an Na⁺ channel blocker, and glibenclamide, apamin, charybdotoxin and XE991, various K⁺ channel blockers, had no effect on clofibrate-induced relaxation. Importantly, clofibrate induced a relaxation response that was not accompanied by any alteration in the cytoplasmic Ca²âº concentration in the longitudinal muscle of the mouse distal colon. Moreover, calyculin A, a myosin light-chain phosphatase (MLCP) inhibitor, attenuated clofibrate-induced relaxation. Our findings indicate that clofibrate relaxes the longitudinal smooth muscle of the mouse distal colon by regulating MLCP activity.


Asunto(s)
Anticolesterolemiantes/farmacología , Calcio/fisiología , Clofibrato/farmacología , Colon/fisiología , Relajación Muscular/fisiología , Músculo Liso/fisiología , Anilidas/farmacología , Animales , Anticolesterolemiantes/metabolismo , Benzamidas/farmacología , Clofibrato/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Guanilato Ciclasa/antagonistas & inhibidores , Masculino , Toxinas Marinas , Ratones , Ratones Endogámicos C57BL , Contracción Muscular/fisiología , Fosfatasa de Miosina de Cadena Ligera/antagonistas & inhibidores , Óxido Nítrico Sintasa/antagonistas & inhibidores , Oxazoles/farmacología , PPAR alfa/agonistas , PPAR alfa/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Piridinas/farmacología , Pirimidinas/farmacología , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Bloqueadores de los Canales de Sodio/farmacología , Guanilil Ciclasa Soluble
18.
Mol Vis ; 17: 1877-90, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21850162

RESUMEN

PURPOSE: The outflow facility for aqueous humor across the trabecular meshwork (TM) is enhanced by agents that oppose the actomyosin contraction of its resident cells. Phosphorylation of MYPT1 (myosin light chain [MLC] phosphatase complex of Type 1) at Thr853 and Thr696 inhibits dephosphorylation of MLC, leading to an increase in actomyosin contraction. In this study, we examined the effects of Rho kinase (ROCK) inhibitors on the relative dephosphorylation of the two sites of MYPT1 using human TM cells (GTM3). METHODS: Dephosphorylation of MYPT1 at Thr853 and Thr696 was determined by western blot analysis following exposure to selective inhibitors of ROCK, namely Y-27632 and Y-39983. Consequent dephosphorylation of MLC and decreases in actomyosin contraction were assessed by western blot analysis and collagen gel contraction assay, respectively. Changes in the cell-matrix adhesion were measured in real time by electric cell-substrate impedance sensing and also assessed by staining for paxillin, vinculin, and focal adhesion kinase (FAK). RESULTS: Both ROCK inhibitors produced a concentration-dependent dephosphorylation at Thr853 and Thr696 of MYPT1 in adherent GTM3 cells. IC50 values for Y-39983 were 15 nM and 177 nM for dephosphorylation at Thr853 and Thr696, respectively. Corresponding values for Y-27632 were 658 nM and 2270 nM. Analysis of the same samples showed a decrease in MLC phosphorylation with IC50 values of 14 nM and 1065 nM for Y-39983 and Y-27632, respectively. Consistent with these changes, both inhibitors opposed contraction of collagen gels induced by TM cells. Exposure of cells to the inhibitors led to a decrease in the electrical cell-substrate resistance, with the effect of Y-39983 being more pronounced than Y-27632. Treatment with these ROCK inhibitors also showed a loss of stress fibers and a concomitant decrease in tyrosine phosphorylation of paxillin and FAK. CONCLUSIONS: Y-39983 and Y-27632 oppose ROCK-dependent phosphorylation of MYPT1 predominantly at Thr853 with a corresponding decrease in MLC phosphorylation. A relatively low effect of both ROCK inhibitors at Thr696 suggests a role for other Ser/Thr kinases at this site. Y-39983 was several-fold more potent when compared with Y-27632 at inhibiting the phosphorylation of MYPT1 at either Thr853 or Thr696 commensurate with its greater potency at inhibiting the activity of human ROCK-I and ROCK-II enzymes.


Asunto(s)
Actomiosina/metabolismo , Uniones Célula-Matriz/metabolismo , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Fosfoproteínas/metabolismo , Fibras de Estrés/efectos de los fármacos , Malla Trabecular , Amidas/farmacología , Humor Acuoso/fisiología , Western Blotting , Línea Celular Transformada , Colágeno/análisis , Proteína-Tirosina Quinasas de Adhesión Focal/análisis , Proteína-Tirosina Quinasas de Adhesión Focal/biosíntesis , Geles/análisis , Humanos , Concentración 50 Inhibidora , Fosfatasa de Miosina de Cadena Ligera/antagonistas & inhibidores , Paxillin/análisis , Paxillin/biosíntesis , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Malla Trabecular/citología , Malla Trabecular/efectos de los fármacos , Malla Trabecular/metabolismo , Vinculina/análisis , Vinculina/biosíntesis
19.
J Thorac Cardiovasc Surg ; 142(2): e59-65, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21397262

RESUMEN

OBJECTIVES: Radial arteries are increasingly used as conduits for coronary artery bypass grafts. However, vasospasm continues to be a major concern in radial artery grafts. Rho kinase plays a critical role in vascular contraction through phosphorylation of the regulatory subunit myosin phosphatase targeting subunit 1 (MYPT1) of the myosin light chain phosphatase to inhibit myosin light chain phosphatase in vascular smooth muscle. The purpose of this study was to evaluate the inhibitory effects of fasudil, a clinically used Rho kinase inhibitor, on Rho kinase activity, myosin light chain phosphorylation, in vitro contraction, and in situ vasospasm in radial arteries of patients undergoing coronary artery bypass grafting surgery. METHODS: The inhibitory efficacy of fasudil on vasoconstrictor-induced contraction and phosphorylation of MYPT1 was examined in radial artery rings. In situ phosphorylation of MYPT1 was evaluated in nonspastic and spastic radial arteries, and the effects of intraluminal administration of fasudil and verapamil-glyceryl trinitrate (VG) on in situ free blood flow and phosphorylation of MYPT1 and myosin light chain were compared in spastic radial arteries. RESULTS: Both fasudil and VG nearly fully inhibited noradrenaline- and serotonin-induced contraction of radial artery rings. However, fasudil but not VG abolished MYPT1 phosphorylation. In spastic radial arteries phosphorylation of MYPT1 and myosin light chain was increased compared with that seen in nonspastic arteries. Intraradial administration of fasudil induced a much larger increase in in situ free blood flow compared with VG treatment. This antispastic effect of fasudil was accompanied by marked decreases in phosphorylation of MYPT1 and myosin light chain. CONCLUSIONS: Fasudil is a very effective Rho kinase inhibitor that deinhibits myosin light chain phosphatase and powerfully relieves vasospasm in situ in radial arteries.


Asunto(s)
1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , Puente de Arteria Coronaria , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , Arteria Radial/efectos de los fármacos , Quinasas Asociadas a rho/antagonistas & inhibidores , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/administración & dosificación , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Anciano , Femenino , Humanos , Inyecciones Intraarteriales , Masculino , Cadenas Ligeras de Miosina/metabolismo , Fosfatasa de Miosina de Cadena Ligera/antagonistas & inhibidores , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Fosforilación/efectos de los fármacos , Arteria Radial/trasplante , Trasplante Autólogo , Vasodilatación , Quinasas Asociadas a rho/metabolismo
20.
Eur J Pharmacol ; 628(1-3): 148-54, 2010 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-19944679

RESUMEN

The present study investigated the mediated effect of protein kinase C (PKC) in arginine vasopressin (AVP)-induced restoration of vascular responsiveness and calcium sensitization following hemorrhagic shock. Using both isolated superior mesenteric artery from hemorrhagic shock rats and hypoxia-treated vascular smooth muscle cell (VSMC), we investigated the roles of PKC-alpha, delta and epsilon isoforms in AVP-induced restoration of vascular reactivity and calcium sensitivity. Meanwhile, effects of their specific inhibitors on the activity of myosin light chain phosphatase (MLCP), myosin light chain kinase (MLCK), and the phosphorylation of myosin light chain (MLC(20)) in VSMC were observed. The results indicated that AVP improved the reactivity of superior mesenteric artery and VSMC to norepinephrine and calcium following hemorrhagic shock and hypoxia. PKC-alpha inhibitor and PKC-epsilon inhibitory peptide antagonized these effects of AVP, while PKC-delta inhibitor only partially antagonized these effects of AVP. AVP up-regulated the expression of PKC-alpha and epsilon in the particulate fractions of hypoxia-treated VSMC with the decrease of the activity of MLCP and the increase of the phosphorylation of MLC(20). These effects of AVP were inhibited by PKC-alpha inhibitor and PKC-epsilon inhibitory peptide, but not by the PKC-delta inhibitor. The results suggested that PKC plays an important role in AVP-induced restoration of vascular reactivity and calcium sensitivity following hemorrhagic shock. PKC-alpha and epsilon may be the main isoforms involved in this process and play effect via MLC(20) phosphorylation dependent mechanism, while PKC-delta may be partially involved in AVP action by other mechanisms.


Asunto(s)
Arginina Vasopresina/farmacología , Calcio/farmacología , Arteria Mesentérica Superior/efectos de los fármacos , Arteria Mesentérica Superior/fisiopatología , Proteína Quinasa C/metabolismo , Recuperación de la Función/efectos de los fármacos , Choque Hemorrágico/fisiopatología , Animales , Hipoxia de la Célula , Células Cultivadas , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Inhibidores Enzimáticos/farmacología , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Técnicas In Vitro , Masculino , Arterias Mesentéricas/citología , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/enzimología , Arterias Mesentéricas/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Fosfatasa de Miosina de Cadena Ligera/antagonistas & inhibidores , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Fosforilación/efectos de los fármacos , Proteína Quinasa C/antagonistas & inhibidores , Ratas , Choque Hemorrágico/metabolismo , Choque Hemorrágico/patología , Vasoconstricción/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...