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










Base de dados
Intervalo de ano de publicação
1.
Cell Biol Int ; 44(9): 1870-1880, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32437058

RESUMO

The objective of this project was to find a bronchodilatory compound from herbs and clarify the mechanism. We found that the ethanol extract of Folium Sennae (EEFS) can relax airway smooth muscle (ASM). EEFS inhibited ASM contraction, induced by acetylcholine, in mouse tracheal rings and lung slices. High-performance liquid chromatography assay showed that EEFS contained emodin. Emodin had a similar reversal action. Acetylcholine-evoked contraction was also partially reduced by nifedipine (a selective inhibitor of L-type voltage-dependent Ca2+ channels, LVDCCs), YM-58483 (a selective inhibitor of store-operated Ca2+ entry, SOCE), as well as Y-27632 (an inhibitor of Rho-associated protein kinase). In addition, LVDCC- and SOCE-mediated currents and cytosolic Ca2+ elevations were inhibited by emodin. Emodin reversed acetylcholine-caused increases in phosphorylation of myosin phosphatase target subunit 1. Furthermore, emodin, in vivo, inhibited acetylcholine-induced respiratory system resistance in mice. These results indicate that EEFS-induced relaxation results from emodin inhibiting LVDCC, SOCE, and Ca2+ sensitization. These findings suggest that Folium Sennae and emodin may be new sources of bronchodilators.


Assuntos
Emodina/farmacologia , Contração Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Acetilcolina/efeitos adversos , Acetilcolina/farmacologia , Animais , Broncodilatadores/metabolismo , Broncodilatadores/farmacologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Contração Muscular/fisiologia , Músculo Liso/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/fisiologia , Extratos Vegetais/farmacologia , Senna/metabolismo
2.
J Biol Chem ; 294(28): 10846-10862, 2019 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-31138649

RESUMO

Cell migration is essential to embryonic development, wound healing, and cancer cell dissemination. Cells move via leading-edge protrusion, substrate adhesion, and retraction of the cell's rear. The molecular mechanisms by which extracellular cues signal to the actomyosin cytoskeleton to control these motility mechanics are poorly understood. The growth factor-responsive and oncogenically activated protein extracellular signal-regulated kinase (ERK) promotes motility by signaling in actin polymerization-mediated edge protrusion. Using a combination of immunoblotting, co-immunoprecipitation, and myosin-binding experiments and cell migration assays, we show here that ERK also signals to the contractile machinery through its substrate, p90 ribosomal S6 kinase (RSK). We probed the signaling and migration dynamics of multiple mammalian cell lines and found that RSK phosphorylates myosin phosphatase-targeting subunit 1 (MYPT1) at Ser-507, which promotes an interaction of Rho kinase (ROCK) with MYPT1 and inhibits myosin targeting. We find that by inhibiting the myosin phosphatase, ERK and RSK promote myosin II-mediated tension for lamella expansion and optimal edge dynamics for cell migration. These findings suggest that ERK activity can coordinately amplify both protrusive and contractile forces for optimal cell motility.


Assuntos
Movimento Celular/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Citoesqueleto de Actina/metabolismo , Actomiosina/metabolismo , Animais , Células COS , Linhagem Celular , Chlorocebus aethiops , Citoesqueleto/metabolismo , Citoesqueleto/fisiologia , Humanos , Contração Muscular , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/fisiologia , Miosinas/metabolismo , Fosforilação , Ligação Proteica , Proteínas Quinases S6 Ribossômicas 90-kDa/fisiologia , Transdução de Sinais , Quinases Associadas a rho/metabolismo
3.
Biochim Biophys Acta Mol Cell Res ; 1866(1): 2-15, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30076859

RESUMO

Myosin phosphatase (MP) holoenzyme is a Ser/Thr specific enzyme, which is the member of protein phosphatase type 1 (PP1) family and composed of a PP1 catalytic subunit (PP1c/PPP1CB) and a myosin phosphatase targeting subunit (MYPT1/PPP1R12A). PP1c is required for the catalytic activity of the holoenzyme, while MYPT1 regulates MP through targeting the holoenzyme to its substrates. Above the well-characterized function of MP, as the major regulator of smooth muscle contractility mediating the dephosphorylation of 20 kDa myosin light chain, accumulating data support its role in other, non-contractile functions. In this review, we summarize the scaffold function of MP holoenzyme and its roles in processes such as cell cycle, development, gene expression regulation and neurotransmitter release. In particular, we highlight novel interacting proteins of MYPT1 and pathophysiological functions of MP relevant to tumorigenesis, insulin resistance and neurodegenerative disorders. This article is part of a Special Issue entitled: Protein Phosphatases as Critical Regulators for Cellular Homeostasis edited by Prof. Peter Ruvolo and Dr. Veerle Janssens.


Assuntos
Fosfatase de Miosina-de-Cadeia-Leve/genética , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/fisiologia , Animais , Humanos , Resistência à Insulina , Miócitos de Músculo Liso/metabolismo , Neoplasias/metabolismo , Neurofibromina 2/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Proteína Fosfatase 1/metabolismo , Proteína Fosfatase 1/fisiologia
4.
J Smooth Muscle Res ; 53(0): 1-19, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28260704

RESUMO

A hallmark of smooth muscle cells is their ability to adapt their functions to meet temporal and chronic fluctuations in their demands. These functions include force development and growth. Understanding the mechanisms underlying the functional plasticity of smooth muscles, the major constituent of organ walls, is fundamental to elucidating pathophysiological rationales of failures of organ functions. Also, the knowledge is expected to facilitate devising innovative strategies that more precisely monitor and normalize organ functions by targeting individual smooth muscles. Evidence has established a current paradigm that the myosin light chain phosphatase (MLCP) is a master regulator of smooth muscle responsiveness to stimuli. Cellular MLCP activity is negatively and positively regulated in response to G-protein activation and cAMP/cGMP production, respectively, through the MYPT1 regulatory subunit and an endogenous inhibitor protein named CPI-17. In this article we review the outcomes from two decade of research on the CPI-17 signaling and discuss emerging paradoxes in the view of signaling pathways regulating smooth muscle functions through MLCP.


Assuntos
Contração Muscular/genética , Músculo Liso/fisiologia , Fosfatase de Miosina-de-Cadeia-Leve/fisiologia , Transdução de Sinais/fisiologia , Cálcio/metabolismo , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Musculares , Tono Muscular/genética , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Fosfoproteínas Fosfatases/fisiologia , Fosforilação , Transdução de Sinais/genética , Quinases Associadas a rho/fisiologia
5.
J Physiol ; 594(12): 3209-25, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-26847850

RESUMO

KEY POINTS: The extent of myosin regulatory light chain phosphorylation (RLC) necessary for smooth muscle contraction depends on the respective activities of Ca(2+) /calmodulin-dependent myosin light chain kinase and myosin light chain phosphatase (MLCP), which contains a regulatory subunit MYPT1 bound to the phosphatase catalytic subunit and myosin. MYPT1 showed significant constitutive T696 and T853 phosphorylation, which is predicted to inhibit MLCP activity in isolated ileal smooth muscle tissues, with additional phosphorylation upon pharmacological treatment with the muscarinic agonist carbachol. Electrical field stimulation (EFS), which releases ACh from nerves, increased force and RLC phosphorylation but not MYPT1 T696 or T853 phosphorylation. The conditional knockout of MYPT1 or the knockin mutation T853A in mice had no effect on the frequency-maximal force responses to EFS in isolated ileal tissues. Physiological RLC phosphorylation and force development in ileal smooth muscle depend on myosin light chain kinase and MLCP activities without changes in constitutive MYPT1 phosphorylation. ABSTRACT: Smooth muscle contraction initiated by myosin regulatory light chain (RLC) phosphorylation is dependent on the relative activities of Ca(2+) /calmodulin-dependent myosin light chain kinase (MLCK) and myosin light chain phosphatase (MLCP). We have investigated the physiological role of the MLCP regulatory subunit MYPT1 in ileal smooth muscle in adult mice with (1) smooth muscle-specific deletion of MYPT1; (2) non-phosphorylatable MYPT1 containing a T853A knockin mutation; and (3) measurements of force and protein phosphorylation responses to cholinergic neurostimulation initiated by electric field stimulation. Isolated MYPT1-deficient tissues from MYPT1(SM-/-) mice contracted and relaxed rapidly with moderate differences in sustained responses to KCl and carbachol treatments and washouts, respectively. Similarly, measurements of regulatory proteins responsible for RLC phosphorylation during contractions also revealed moderate changes. There were no differences in contractile or RLC phosphorylation responses to carbachol between tissues from normal mice vs. MYPT1 T853A knockin mice. Quantitatively, there was substantial MYPT1 T696 and T853 phosphorylation in wild-type tissues under resting conditions, predicting a high extent of MLCP phosphatase inhibition. Reduced PP1cδ activity in MYPT1-deficient tissues may be similar to attenuated MLCP activity in wild-type tissues resulting from constitutively phosphorylated MYPT1. Electric field stimulation increased RLC phosphorylation and force development in tissues from wild-type mice without an increase in MYPT1 phosphorylation. Thus, physiological RLC phosphorylation and force development in ileal smooth muscle appear to be dependent on MLCK and MLCP activities without changes in constitutive MYPT1 phosphorylation.


Assuntos
Íleo/fisiologia , Músculo Liso/fisiologia , Fosfatase de Miosina-de-Cadeia-Leve/fisiologia , Animais , Carbacol/farmacologia , Estimulação Elétrica , Íleo/metabolismo , Íleo/patologia , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos Transgênicos , Contração Muscular/efeitos dos fármacos , Proteínas Musculares/metabolismo , Músculo Liso/metabolismo , Músculo Liso/patologia , Cadeias Leves de Miosina/metabolismo , Cadeias Leves de Miosina/fisiologia , Quinase de Cadeia Leve de Miosina/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/genética , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Fosfoproteínas/metabolismo , Fosforilação , Cloreto de Potássio/farmacologia , Transdução de Sinais
6.
Circ Res ; 116(5): 895-908, 2015 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-25722443

RESUMO

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.


Assuntos
Doenças Cardiovasculares/fisiopatologia , Endotélio Vascular/fisiopatologia , Mecanotransdução Celular/fisiologia , Rigidez Vascular/fisiologia , Quinases Associadas a rho/fisiologia , Actomiosina/fisiologia , Envelhecimento/fisiologia , Animais , Calcinose/patologia , Calcinose/fisiopatologia , Doenças Cardiovasculares/enzimologia , Adesão Celular/fisiologia , Permeabilidade da Membrana Celular , Citoesqueleto/ultraestrutura , Endotélio Vascular/ultraestrutura , Hemorreologia , Humanos , Inflamação , Integrinas/fisiologia , Leucócitos/fisiologia , Mecanotransdução Celular/efeitos dos fármacos , Camundongos , Camundongos Knockout , Microcirculação , Modelos Cardiovasculares , Fosfatase de Miosina-de-Cadeia-Leve/antagonistas & inibidores , Fosfatase de Miosina-de-Cadeia-Leve/fisiologia , NF-kappa B/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Análise de Onda de Pulso , Ratos , Migração Transendotelial e Transepitelial , Rigidez Vascular/efeitos dos fármacos , Proteínas rho de Ligação ao GTP/antagonistas & inibidores , Proteínas rho de Ligação ao GTP/fisiologia , Quinases Associadas a rho/antagonistas & inibidores
7.
J Physiol ; 592(14): 3031-51, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24835173

RESUMO

Smooth muscle contraction initiated by myosin regulatory light chain (RLC) phosphorylation is dependent on the relative activities of Ca(2+)-calmodulin-dependent myosin light chain kinase (MLCK) and myosin light chain phosphatase (MLCP). We have investigated the physiological role of the MLCP regulatory subunit MYPT1 in bladder smooth muscle containing a smooth muscle-specific deletion of MYPT1 in adult mice. Deep-sequencing analyses of mRNA and immunoblotting revealed that MYPT1 depletion reduced the amount of PP1cδ with no compensatory changes in expression of other MYPT1 family members. Phosphatase activity towards phosphorylated smooth muscle heavy meromyosin was proportional to the amount of PP1cδ in total homogenates from wild-type or MYPT1-deficient tissues. Isolated MYPT1-deficient tissues from MYPT1(SM-/-) mice contracted with moderate differences in response to KCl and carbachol treatments, and relaxed rapidly with comparable rates after carbachol removal and only 1.5-fold slower after KCl removal. Measurements of phosphorylated proteins in the RLC signalling and actin polymerization modules during contractions revealed moderate changes. Using a novel procedure to quantify total phosphorylation of MYPT1 at Thr696 and Thr853, we found substantial phosphorylation in wild-type tissues under resting conditions, predicting attenuation of MLCP activity. Reduced PP1cδ activity in MYPT1-deficient tissues may be similar to the attenuated MLCP activity in wild-type tissues resulting from constitutively phosphorylated MYPT1. Constitutive phosphorylation of MYPT1 Thr696 and Thr853 may thus represent a physiological mechanism acting in concert with agonist-induced MYPT1 phosphorylation to inhibit MLCP activity. In summary, MYPT1 deficiency may not cause significant derangement of smooth muscle contractility because the effective MLCP activity is not changed.


Assuntos
Músculo Liso/fisiologia , Quinase de Cadeia Leve de Miosina/fisiologia , Fosfatase de Miosina-de-Cadeia-Leve/fisiologia , Bexiga Urinária/fisiologia , Animais , Sequenciamento de Nucleotídeos em Larga Escala , Masculino , Camundongos Transgênicos , Contração Muscular , Fosforilação , RNA Mensageiro/genética
8.
J Smooth Muscle Res ; 50: 18-28, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24770446

RESUMO

Smooth muscle contraction is activated primarily by phosphorylation at Ser19 of the regulatory light chain subunits (LC20) of myosin II, catalysed by Ca(2+)/calmodulin-dependent myosin light chain kinase. Ca(2+)-independent contraction can be induced by inhibition of myosin light chain phosphatase, which correlates with diphosphorylation of LC20 at Ser19 and Thr18, catalysed by integrin-linked kinase (ILK) and zipper-interacting protein kinase (ZIPK). LC20 diphosphorylation at Ser19 and Thr18 has been detected in mammalian vascular smooth muscle tissues in response to specific contractile stimuli (e.g. endothelin-1 stimulation of rat renal afferent arterioles) and in pathophysiological situations associated with hypercontractility (e.g. cerebral vasospasm following subarachnoid hemorrhage). Comparison of the effects of LC 20 monophosphorylation at Ser19 and diphosphorylation at Ser19 and Thr18 on contraction and relaxation of Triton-skinned rat caudal arterial smooth muscle revealed that phosphorylation at Thr18 has no effect on steady-state force induced by Ser19 phosphorylation. On the other hand, the rates of dephosphorylation and relaxation are significantly slower following diphosphorylation at Thr18 and Ser19 compared to monophosphorylation at Ser19. We propose that this diphosphorylation mechanism underlies the prolonged contractile response of particular vascular smooth muscle tissues to specific stimuli, e.g. endothelin-1 stimulation of renal afferent arterioles, and the vasospastic behavior observed in pathological conditions such as cerebral vasospasm following subarachnoid hemorrhage and coronary arterial vasospasm. ILK and ZIPK may, therefore, be useful therapeutic targets for the treatment of such conditions.


Assuntos
Músculo Liso Vascular/fisiologia , Miosina Tipo II/química , Miosina Tipo II/fisiologia , Vasoconstrição/genética , Injúria Renal Aguda/tratamento farmacológico , Animais , Catálise , Vasoespasmo Coronário/tratamento farmacológico , Proteínas Quinases Associadas com Morte Celular/fisiologia , Proteínas Quinases Associadas com Morte Celular/uso terapêutico , Endotelina-1/farmacologia , Humanos , Hipertensão/tratamento farmacológico , Microcirculação/efeitos dos fármacos , Microcirculação/genética , Terapia de Alvo Molecular , Quinase de Cadeia Leve de Miosina/fisiologia , Fosfatase de Miosina-de-Cadeia-Leve/fisiologia , Fosforilação , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Serina-Treonina Quinases/uso terapêutico , Ratos , Circulação Renal/efeitos dos fármacos , Circulação Renal/genética , Vasoespasmo Intracraniano/tratamento farmacológico
9.
PLoS One ; 9(4): e94684, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24732978

RESUMO

Cell shape changes are crucial for metazoan development. During Caenorhabditis elegans embryogenesis, epidermal cell shape changes transform ovoid embryos into vermiform larvae. This process is divided into two phases: early and late elongation. Early elongation involves the contraction of filamentous actin bundles by phosphorylated non-muscle myosin in a subset of epidermal (hypodermal) cells. The genes controlling early elongation are associated with two parallel pathways. The first one involves the rho-1/RHOA-specific effector let-502/Rho-kinase and mel-11/myosin phosphatase regulatory subunit. The second pathway involves the CDC42/RAC-specific effector pak-1. Late elongation is driven by mechanotransduction in ventral and dorsal hypodermal cells in response to body-wall muscle contractions, and involves the CDC42/RAC-specific Guanine-nucleotide Exchange Factor (GEF) pix-1, the GTPase ced-10/RAC and pak-1. In this study, pix-1 is shown to control early elongation in parallel with let-502/mel-11, as previously shown for pak-1. We show that pix-1, pak-1 and let-502 control the rate of elongation, and the antero-posterior morphology of the embryos. In particular, pix-1 and pak-1 are shown to control head, but not tail width, while let-502 controls both head and tail width. This suggests that let-502 function is required throughout the antero-posterior axis of the embryo during early elongation, while pix-1/pak-1 function may be mostly required in the anterior part of the embryo. Supporting this hypothesis we show that low pix-1 expression level in the dorsal-posterior hypodermal cells is required to ensure high elongation rate during early elongation.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/genética , Proteínas de Transporte/fisiologia , Fosfatase de Miosina-de-Cadeia-Leve/fisiologia , Quinases Associadas a rho/fisiologia , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/metabolismo , Citoplasma/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Mecanotransdução Celular/genética , Mutação , Fenótipo , Fosforilação , Transdução de Sinais
10.
Blood ; 122(20): 3533-45, 2013 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-24100445

RESUMO

Cyclic adenosine monophosphate (cAMP)-dependent signaling modulates platelet shape change through unknown mechanisms. We examined the effects of cAMP signaling on platelet contractile machinery. Prostaglandin E1 (PGE1)-mediated inhibition of thrombin-stimulated shape change was accompanied by diminished phosphorylation of myosin light chain (MLC). Since thrombin stimulates phospho-MLC through RhoA/Rho-associated, coiled-coil containing protein kinase (ROCK)-dependent inhibition of MLC phosphatase (MLCP), we examined the effects of cAMP on this pathway. Thrombin stimulated the membrane localization of RhoA and the formation of a signaling complex of RhoA/ROCK2/myosin phosphatase-targeting subunit 1 (MYPT1). This resulted in ROCK-mediated phosphorylation of MYPT1 on threonine 853 (thr(853)), the disassociation of the catalytic subunit protein phosphatase 1δ (PP1δ) from MYPT1 and inhibition of basal MLCP activity. Treatment of platelets with PGE1 prevented thrombin-induced phospho-MYPT1-thr(853) in a protein kinase A (PKA)-dependent manner. Examination of the molecular mechanisms revealed that PGE1 induced the phosphorylation of RhoA on serine(188) through a pathway requiring cAMP and PKA. This event inhibited the membrane relocalization of RhoA, prevented the association of RhoA with ROCK2 and MYPT1, attenuated the dissociation of PP1δ from MYPT1, and thereby restored basal MLCP activity leading to a decrease in phospho-MLC. These data reveal a new mechanism by which the cAMP-PKA signaling pathway regulates platelet function.


Assuntos
Plaquetas/enzimologia , AMP Cíclico/fisiologia , Quinase de Cadeia Leve de Miosina/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Sistemas do Segundo Mensageiro/fisiologia , Transdução de Sinais/fisiologia , Quinases Associadas a rho/fisiologia , Proteína rhoA de Ligação ao GTP/fisiologia , Alprostadil/farmacologia , Plaquetas/efeitos dos fármacos , Plaquetas/ultraestrutura , Forma Celular/efeitos dos fármacos , Forma Celular/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Humanos , Técnicas In Vitro , Complexos Multiproteicos , Quinase de Cadeia Leve de Miosina/sangue , Fosforilação , Fosfosserina/metabolismo , Fosfotreonina/metabolismo , Proteína Fosfatase 1/metabolismo , Subunidades Proteicas , Trombina/farmacologia
11.
Circ Res ; 112(2): 257-66, 2013 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-23168335

RESUMO

RATIONALE: Nkx2.5 is a transcription factor that regulates cardiomyogenesis in vivo and in embryonic stem cells. It is also a common target in congenital heart disease. Although Nkx2.5 has been implicated in the regulation of many cellular processes that ultimately contribute to cardiomyogenesis and morphogenesis of the mature heart, relatively little is known about how it is regulated at a functional level. OBJECTIVE: We have undertaken a proteomic screen to identify novel binding partners of Nkx2.5 during cardiomyogenic differentiation in an effort to better understand the regulation of its transcriptional activity. METHODS AND RESULTS: Purification of Nkx2.5 from differentiating cells identified the myosin phosphatase subunits protein phosphatase 1ß and myosin phosphatase targeting subunit 1 (Mypt1) as novel binding partners. The interaction with protein phosphatase 1 ß/Mypt1 resulted in exclusion of Nkx2.5 from the nucleus and, consequently, inhibition of its transcriptional activity. Exclusion of Nkx2.5 was inhibited by treatment with leptomycin B and was dependent on an Mypt1 nuclear export signal. Furthermore, in transient transfection experiments, Nkx2.5 colocalized outside the nucleus with phosphorylated Mypt1 in a manner dependent on Wnt signaling and Rho-associated protein kinase. Treatment of differentiating mouse embryonic stem cells with Wnt3a resulted in enhanced phosphorylation of endogenous Mypt1, increased nuclear exclusion of endogenous Nkx2.5, and a failure to undergo terminal cardiomyogenesis. Finally, knockdown of Mypt1 resulted in rescue of Wnt3a-mediated inhibition of cardiomyogenesis, indicating that Mypt1 is required for this process. CONCLUSIONS: We have identified a novel interaction between Nkx2.5 and myosin phosphatase. Promoting this interaction represents a novel mechanism whereby Wnt3a regulates Nkx2.5 and inhibits cardiomyogenesis.


Assuntos
Inibidores do Crescimento/fisiologia , Proteínas de Homeodomínio/metabolismo , Miócitos Cardíacos/fisiologia , Fosfatase de Miosina-de-Cadeia-Leve/fisiologia , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Via de Sinalização Wnt/fisiologia , Proteína Wnt3A/fisiologia , Quinases Associadas a rho/fisiologia , Animais , Células-Tronco Embrionárias/enzimologia , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/fisiologia , Células HEK293 , Proteína Homeobox Nkx-2.5 , Humanos , Camundongos , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Proteína Fosfatase 1/metabolismo , Frações Subcelulares/enzimologia , Frações Subcelulares/metabolismo
12.
Pflugers Arch ; 463(2): 257-68, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22020732

RESUMO

The present study was to determine the role of the type I isoform of cGMP-dependent protein kinase (PKG I) and its downstream effector myosin phosphatase target subunit 1 (MYPT1) in the responses of different sized coronary arteries to nitrovasodilators. Relaxations of isolated porcine coronary arteries were determined by isometric tension recording technique. Protein levels of PKG I and its effectors were analyzed by Western blotting. The activities of PKG I and MYPT1 were studied by analyzing phosphorylation of vasodilator-stimulated phosphoprotein (VASP) and MYPT1, respectively. Nitroglycerin, DETA NONOate, and 8-Br-cGMP caused greater relaxations in large than in small coronary arteries. Relaxations were attenuated to a greater extent by Rp-8-Br-PET-cGMPS (a PKG inhibitor) in large vs. small arteries. The expressions of PKG I and MYPT1 in large arteries were more abundant than in small arteries. DETA NONOate stimulated phosphorylation of VASP at Ser239 and inhibited phosphorylation of MYPT1 at Thr853 to a greater extent in large than in small arteries. A suppressed phosphorylation of MYPT1 at Thr853 was caused by 8-Br-cGMP in large but not small arteries, which was inhibited by Rp-8-Br-PET-cGMPS. These results suggest that the greater responsiveness of large coronary arteries to nitrovasodilators result in part from greater activities of PKG I and MYPT1. Dysfunction in nitric oxide signaling is implicated in the vulnerability of large coronary arteries to certain disorders such as atherosclerosis and spasm. Augmentation of PKG I-MYPT1 signaling may be of therapeutic benefit for combating these events.


Assuntos
Vasos Coronários/patologia , Vasos Coronários/fisiologia , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Fosfatase de Miosina-de-Cadeia-Leve/fisiologia , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Animais , Moléculas de Adesão Celular/metabolismo , Vasos Coronários/efeitos dos fármacos , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Proteínas dos Microfilamentos/metabolismo , Modelos Animais , Óxido Nítrico/fisiologia , Nitroglicerina/farmacologia , Compostos Nitrosos/farmacologia , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Transdução de Sinais/fisiologia , Suínos , Vasodilatação/fisiologia
13.
Cell Mol Life Sci ; 69(2): 247-66, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21947498

RESUMO

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.


Assuntos
Músculo Liso Vascular/efeitos dos fármacos , Nucleotídeos Cíclicos/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Animais , Cálcio/metabolismo , Cálcio/fisiologia , ATPases Transportadoras de Cálcio/metabolismo , ATPases Transportadoras de Cálcio/fisiologia , Humanos , Camundongos , Contração Muscular/efeitos dos fármacos , Contração Muscular/fisiologia , Músculo Liso Vascular/fisiologia , Quinase de Cadeia Leve de Miosina/antagonistas & inibidores , Quinase de Cadeia Leve de Miosina/metabolismo , Quinase de Cadeia Leve de Miosina/fisiologia , Fosfatase de Miosina-de-Cadeia-Leve/antagonistas & inibidores , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/fisiologia , Nucleotídeos Cíclicos/metabolismo , Nucleotídeos Cíclicos/fisiologia , Canais de Potássio/agonistas , Canais de Potássio/metabolismo , Canais de Potássio/fisiologia , Ratos , Retículo Sarcoplasmático/efeitos dos fármacos , Retículo Sarcoplasmático/metabolismo , Retículo Sarcoplasmático/fisiologia , Trocador de Sódio e Cálcio/metabolismo , Trocador de Sódio e Cálcio/fisiologia , Vasodilatação/fisiologia , Vasodilatadores/metabolismo
14.
Hum Reprod Update ; 16(6): 725-44, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20551073

RESUMO

BACKGROUND: Uterine contractile activity plays an important role in many and varied reproductive functions including sperm and embryo transport, implantation, menstruation, gestation and parturition. Abnormal contractility might underlie common and important disorders such as infertility, implantation failure, dysmenorrhea, endometriosis, spontaneous miscarriage or preterm birth. METHODS: A systematic review of the US National Library of Medicine was performed linking 'uterus' or 'uterine myocyte' with 'calcium ion' (Ca(2+)), 'myosin light chain kinase' and 'myosin light chain phosphatase'. This led to many cross-references involving non-uterine myocytes and, where relevant, these data have been incorporated into the following synthesis. RESULTS: We have grouped the data according to three main components that determine uterine contractility: the contractile apparatus; electrophysiology of the myocyte including excitation-contraction coupling; and regulation of the sensitivity of the contractile apparatus to Ca(2+). We also have included information regarding potential therapeutic methods for regulating uterine contractility. CONCLUSIONS: More research is necessary to understand the mechanisms that generate the frequency, amplitude, duration and direction of propagation of uterine contractile activity. On the basis of current knowledge of the molecular control of uterine myocyte function, there are opportunities for systematic testing of the efficacy of a variety of available potential pharmacological agents and for the development of new agents. Taking advantage of these opportunities could result in an overall improvement in reproductive health.


Assuntos
Contração Uterina/fisiologia , Citoesqueleto de Actina/fisiologia , Sinalização do Cálcio , Eletrofisiologia , Feminino , Humanos , Modelos Biológicos , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/fisiologia , Miócitos de Músculo Liso/ultraestrutura , Miométrio/anatomia & histologia , Miométrio/metabolismo , Miométrio/fisiologia , Quinase de Cadeia Leve de Miosina/metabolismo , Quinase de Cadeia Leve de Miosina/fisiologia , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/fisiologia , Contração Uterina/metabolismo , Quinases Associadas a rho/metabolismo , Quinases Associadas a rho/fisiologia , Proteína rhoA de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/fisiologia
15.
Oncogene ; 29(29): 4183-93, 2010 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-20498637

RESUMO

Cancer cells often have unstable genomes and increased centrosome and chromosome numbers, which are an important part of malignant transformation in the most recent model of tumorigenesis. However, very little is known about divisional failures in cancer cells that may lead to chromosomal and centrosomal amplifications. In this study, we show that cancer cells often failed at cytokinesis because of decreased phosphorylation of the myosin regulatory light chain (MLC), a key regulatory component of cortical contraction during division. Reduced MLC phosphorylation was associated with high expression of myosin phosphatase and/or reduced myosin light-chain kinase levels. Furthermore, expression of phosphomimetic MLC largely prevented cytokinesis failure in the tested cancer cells. When myosin light-chain phosphorylation was restored to normal levels by phosphatase knockdown, multinucleation and multipolar mitosis were markedly reduced, resulting in enhanced genome stabilization. Furthermore, both overexpression of myosin phosphatase or inhibition of the myosin light-chain kinase in nonmalignant cells could recapitulate some of the mitotic defects of cancer cells, including multinucleation and multipolar spindles, indicating that these changes are sufficient to reproduce the cytokinesis failures we see in cancer cells. These results for the first time define the molecular defects leading to divisional failure in cancer cells.


Assuntos
Citocinese , Cadeias Leves de Miosina/metabolismo , Neoplasias/patologia , Linhagem Celular Tumoral , Polaridade Celular , Humanos , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , Quinase de Cadeia Leve de Miosina/antagonistas & inibidores , Fosfatase de Miosina-de-Cadeia-Leve/análise , Fosfatase de Miosina-de-Cadeia-Leve/fisiologia , Neoplasias/metabolismo , Fosforilação
16.
J Cell Biochem ; 111(2): 362-9, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20506308

RESUMO

Both rat derived vascular smooth muscle cells (SMC) and human myofibroblasts contain α smooth muscle actin (SMA), but they utilize different mechanisms to contract populated collagen lattices (PCLs). The difference is in how the cells generate the force that contracts the lattices. Human dermal fibroblasts transform into myofibroblasts, expressing α-SMA within stress fibers, when cultured in lattices that remain attached to the surface of a tissue culture dish. When attached lattices are populated with rat derived vascular SMC, the cells retain their vascular SMC phenotype. Comparing the contraction of attached PCLs when they are released from the culture dish on day 4 shows that lattices populated with rat vascular SMC contract less than those populated with human myofibroblast. PCL contraction was evaluated in the presence of vanadate and genistein, which modify protein tyrosine phosphorylation, and ML-7 and Y-27632, which modify myosin ATPase activity. Genistein and ML-7 had no affect upon either myofibroblast or vascular SMC-PCL contraction, demonstrating that neither protein tyrosine kinase nor myosin light chain kinase was involved. Vanadate inhibited myofibroblast-PCL contraction, consistent with a role for protein tyrosine phosphatase activity with myofibroblast-generated forces. Y-27632 inhibited both SMC and myofibroblast PCL contraction, consistent with a central role of myosin light chain phosphatase.


Assuntos
Colágeno/fisiologia , Contração Muscular , Miócitos de Músculo Liso/fisiologia , Miofibroblastos/fisiologia , Animais , Fenômenos Biomecânicos , Células Cultivadas , Humanos , Fosfatase de Miosina-de-Cadeia-Leve/fisiologia , Proteínas Tirosina Fosfatases/fisiologia , Ratos , Especificidade da Espécie
17.
Development ; 137(5): 795-804, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20147380

RESUMO

We demonstrate that in the zebrafish hindbrain, cell shape, rhombomere morphogenesis and, unexpectedly, brain ventricle lumen expansion depend on the contractile state of the neuroepithelium. The hindbrain neural tube opens in a specific sequence, with initial separation along the midline at rhombomere boundaries, subsequent openings within rhombomeres and eventual coalescence of openings into the hindbrain ventricle lumen. A mutation in the myosin phosphatase regulator mypt1 results in a small ventricle due to impaired stretching of the surrounding neuroepithelium. Although initial hindbrain opening remains normal, mypt1 mutant rhombomeres do not undergo normal morphological progression. Three-dimensional reconstruction demonstrates cell shapes within rhombomeres and at rhombomere boundaries are abnormal in mypt1 mutants. Wild-type cell shape requires that surrounding cells are also wild type, whereas mutant cell shape is autonomously regulated. Supporting the requirement for regulation of myosin function during hindbrain morphogenesis, wild-type embryos show dynamic levels of phosphorylated myosin regulatory light chain (pMRLC). By contrast, mutants show continuously high pMRLC levels, with concentration of pMRLC and myosin II at the apical side of the epithelium, and myosin II and actin concentration at rhombomere boundaries. Brain ventricle lumen expansion, rhombomere morphology and cell shape are rescued by inhibition of myosin II function, indicating that each defect is a consequence of overactive myosin. We suggest that the epithelium must ;relax', via activity of myosin phosphatase, to allow for normal hindbrain morphogenesis and expansion of the brain ventricular lumen. Epithelial relaxation might be a widespread strategy to facilitate tube inflation in many organs.


Assuntos
Ventrículos Cerebrais/embriologia , Células Epiteliais/fisiologia , Fosfatase de Miosina-de-Cadeia-Leve/fisiologia , Rombencéfalo/embriologia , Animais , Animais Geneticamente Modificados , Padronização Corporal/genética , Padronização Corporal/fisiologia , Movimento Celular/genética , Movimento Celular/fisiologia , Forma Celular/genética , Ventrículos Cerebrais/metabolismo , Embrião não Mamífero , Células Epiteliais/metabolismo , Modelos Biológicos , Morfogênese/fisiologia , Fosfatase de Miosina-de-Cadeia-Leve/genética , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Rombencéfalo/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/fisiologia
18.
Crit Care Med ; 38(3): 861-70, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20168148

RESUMO

OBJECTIVE: To investigate the molecular mechanisms leading to edema-induced decreases in intestinal smooth muscle myosin light-chain phosphorylation. Intestinal interstitial edema often develops during abdominal surgery and after fluid resuscitation in trauma patients. Intestinal edema causes decreased intestinal contractile activity via decreased intestinal smooth muscle myosin light-chain phosphorylation, leading to slower intestinal transit. Interstitial edema development is a complex phenomenon, resulting in many changes to the interstitial environment surrounding intestinal smooth muscle cells. Thus, the mechanism(s) by which intestinal edema development causes intestinal dysfunction are likely to be multifactorial. DESIGN: Randomized animal study. SETTING: University laboratory. SUBJECTS: Male Sprague-Dawley rats, weighing 250-350 g. INTERVENTION: Studies were performed in a rat model in which a combination of mesenteric venous hypertension and administration of resuscitative fluids induces intestinal edema, mimicking the clinical setting of damage control resuscitation. MEASUREMENTS AND MAIN RESULTS: Microarray analysis of edematous intestinal smooth muscle combined with an in silico search for overrepresented transcription factor binding sites revealed the involvement of nuclear factor-kappaB in edema-induced intestinal dysfunction. Nuclear factor-kappaB deoxyribonucleic acid binding activity was significantly increased in edematous intestinal smooth muscle compared with controls. Inhibition of nuclear factor-kappaB activation blocked edema-induced decreases in basal intestinal contractile activity. Inhibition of nuclear factor-kappaB activation also attenuated edema-induced decreases in myosin light-chain phosphorylation. CONCLUSIONS: We conclude that intestinal edema activates nuclear factor-kappaB, which, in turn, triggers a gene regulation program that eventually leads to decreased myosin light-chain phosphorylation and, thus, decreased intestinal contractile activity.


Assuntos
Edema/fisiopatologia , Motilidade Gastrointestinal/fisiologia , Enteropatias/fisiopatologia , Fosfatase de Miosina-de-Cadeia-Leve/fisiologia , NF-kappa B/fisiologia , Transdução de Sinais/fisiologia , Animais , Hidratação , Regulação da Expressão Gênica/fisiologia , Masculino , Músculo Liso/fisiopatologia , Fosfatase de Miosina-de-Cadeia-Leve/genética , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação/genética , Fosforilação/fisiologia , Ratos , Ratos Sprague-Dawley , Ressuscitação , Transdução de Sinais/genética , Pressão Venosa/fisiologia
19.
Curr Eye Res ; 34(3): 177-83, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19274524

RESUMO

PURPOSE: The purpose of the present study was to clarify the role of Rho-kinase and/or protein kinase C in the resting tension of the isolated pig iris sphincter muscle. MATERIALS AND METHODS: The motor activity of the isolated pig iris sphincter muscle was measured isometrically. RESULTS: EGTA, a chelator of extracellular Ca(2+), significantly reduced the resting tension. Y27632, a Rho-kinase inhibitor, significantly reduced the resting tension in a concentration-dependent manner. The resting tension diminished by Y27632 was significantly recovered by the addition of calyculin A, a myosin light chain phosphatase (MLCP) inhibitor, in a concentration-dependent manner. GF109203X, a protein kinase C inhibitor, had no effect on the resting tension. CONCLUSION: These results suggest that, in the isolated pig iris sphincter muscle, Rho-kinase plays an important role in the generation of spontaneous tone in the resting phase via the inhibition of MLCP activity.


Assuntos
Iris/enzimologia , Músculo Liso/fisiologia , Proteína Quinase C/fisiologia , Quinases Associadas a rho/fisiologia , Animais , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Contração Muscular/efeitos dos fármacos , Contração Muscular/fisiologia , Músculo Liso/inervação , Fosfatase de Miosina-de-Cadeia-Leve/antagonistas & inibidores , Fosfatase de Miosina-de-Cadeia-Leve/fisiologia , Sistema Nervoso Parassimpático/fisiologia , Proteína Quinase C/antagonistas & inibidores , Pupila/fisiologia , Suínos , Quinases Associadas a rho/antagonistas & inibidores
20.
Eur J Pharmacol ; 590(1-3): 61-6, 2008 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-18579130

RESUMO

H89 (N-[2-(p-bromocinnamylamino)ethyl]-5-isoquinolinesulfonamide) is a compound characterized in vitro as a potent and selective inhibitor of protein kinase A (PKA). In this study, we found that H89 reduced the phosphorylation of the myosin regulatory light chain (MRLC) at Thr-18/Ser-19 and induced disassembly of stress fibers in HeLa cells. In addition, we found that H89 induced not only reduction of the MRLC phosphorylation but also cell growth inhibition in several human cancer cell lines. Recently H89 has been found to inhibit Rho-kinase with potency similar to or greater than that for inhibition of PKA. Indeed, the effects of H89 on both the MRLC phosphorylation and actin cytoskeleton organization were nearly identical to those of Rho-kinase inhibitor Y-27632. However, unlike H89, Y-27632 did not affect cell growth of HeLa cells. Further, when the myosin phosphatase targeting subunit 1 (MYPT1) expression was silenced by RNA interference in HeLa cells, the suppressive effect of H89 on the MRLC phosphorylation was not affected, while H89-induced cell growth inhibition was blocked. These results suggest that H89-induced reduction of the MRLC phosphorylation results from inhibition of Rho-kinase and that H89-induced cell growth inhibition is independent of reduction of the MRLC phosphorylation.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Isoquinolinas/farmacologia , Cadeias Leves de Miosina/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Sulfonamidas/farmacologia , Actinas/química , Amidas/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Citoesqueleto/efeitos dos fármacos , Humanos , Fosfatase de Miosina-de-Cadeia-Leve/fisiologia , Fosforilação , Piridinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...