Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 169
Filtrar
1.
Front Immunol ; 12: 697071, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34745088

RESUMO

Background: High mobility group box 1 (HMGB1) causes microvascular endothelial cell barrier dysfunction during acute lung injury (ALI) in sepsis, but the mechanisms have not been well understood. We studied the roles of RAGE and Rho kinase 1 (ROCK1) in HMGB1-induced human pulmonary endothelial barrier disruption. Methods: In the present study, the recombinant human high mobility group box 1 (rhHMGB1) was used to stimulate human pulmonary microvascular endothelial cells (HPMECs). The endothelial cell (EC) barrier permeability was examined by detecting FITC-dextran flux. CCK-8 assay was used to detect cell viability under rhHMGB1 treatments. The expression of related molecules involved in RhoA/ROCK1 pathway, phosphorylation of myosin light chain (MLC), F-actin, VE-cadherin and ZO-1 of different treated groups were measured by pull-down assay, western blot and immunofluorescence. Furthermore, we studied the effects of Rho kinase inhibitor (Y-27632), ROCK1/2 siRNA, RAGE-specific blocker (FPS-ZM1) and RAGE siRNA on endothelial barrier properties to elucidate the related mechanisms. Results: In the present study, we demonstrated that rhHMGB1 induced EC barrier hyperpermeability in a dose-dependent and time-dependent manner by measuring FITC-dextran flux, a reflection of the loss of EC barrier integrity. Moreover, rhHMGB1 induced a dose-dependent and time-dependent increases in paracellular gap formation accompanied by the development of stress fiber rearrangement and disruption of VE-cadherin and ZO-1, a phenotypic change related to increased endothelial contractility and endothelial barrier permeability. Using inhibitors and siRNAs directed against RAGE and ROCK1/2, we systematically determined that RAGE mediated the rhHMGB1-induced stress fiber reorganization via RhoA/ROCK1 signaling activation and the subsequent MLC phosphorylation in ECs. Conclusion: HMGB1 is capable of disrupting the endothelial barrier integrity. This study demonstrates that HMGB1 activates RhoA/ROCK1 pathway via RAGE, which phosphorylates MLC inducing stress fiber formation at short time, and HMGB1/RAGE reduces AJ/TJ expression at long term independently of RhoA/ROCK1 signaling pathway.


Assuntos
Permeabilidade Capilar/fisiologia , Células Endoteliais/metabolismo , Proteína HMGB1/fisiologia , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Quinases Associadas a rho/fisiologia , Células Cultivadas , Humanos , Cadeias Leves de Miosina/fisiologia , Transdução de Sinais/fisiologia
2.
Clin Res Hepatol Gastroenterol ; 44(1): 101-114, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31255531

RESUMO

BACKGROUND/AIMS: Colon cancer is a common cancer that is a threat to human health. Some long non-coding RNAs (lncRNAs) have been observed to exert roles in colon cancer. Here, the current study is aimed to explore the potential mechanism of lncRNA MBNL1 antisense RNA 1 (MBNL1-AS1) in progression of colon cancer and the associated mechanisms. METHODS: Microarray analysis was performed to screen differentially expressed lncRNA and genes associated with colon cancer and its potential mechanism. The functional role of MBNL1-AS1 in colon cancer was analyzed, followed identification of the interaction among MBNL1-AS1, microRNA-412-3p (miR-412-3p), and MYL9. Subsequently, CSC viability, migration, invasion, and apoptosis were detected though a series of in vitro experiments. At last, in vivo experiments were performed to assess tumor formation of colon CSCs. RESULTS: MBNL1-AS1 and MYL9 were poorly expressed in colon cancer. MBNL1-AS1 could competitively bind to miR-412-3p so as to promote MYL9 expression. Enhancement of MBNL1-AS1 or inhibition of miR-412-3p was shown to decrease CSC proliferation, migration, and invasion but promote apoptosis. Moreover, MBNL1-AS1 reversed the CSC-like properties as well as xenograft tumor formation in vivo induced by miR-412-3p. CONCLUSION: Collectively, the present study suggests an inhibitory role of MBNL1-AS1 in colon cancer by upregulating miR-412-3p-targeted MYL9. Thus, this study provides an enhanced understanding of MBNL1-AS1 along with miR-412-3p and MYL9 as therapeutic targets for colon cancer.


Assuntos
Movimento Celular , Proliferação de Células , Neoplasias do Colo/patologia , MicroRNAs/fisiologia , Cadeias Leves de Miosina/fisiologia , Células-Tronco Neoplásicas/fisiologia , Proteínas de Ligação a RNA/fisiologia , Humanos , Invasividade Neoplásica , RNA Longo não Codificante , Proteínas de Ligação a RNA/genética , Células Tumorais Cultivadas
3.
Am J Physiol Cell Physiol ; 316(4): C509-C521, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30698990

RESUMO

The phosphorylation state of myosin regulatory light chain (MRLC) is central to the regulation of contractility that impacts cellular homeostasis and fate decisions. Rho-kinase (ROCK) and myosin light chain kinase (MLCK) are major kinases for MRLC documented to selectively regulate MRLC in a subcellular position-specific manner; specifically, MLCK in some nonmuscle cell types works in the cell periphery to promote migration, while ROCK does so at the central region to sustain contractility. However, it remains unclear whether or not the spatially selective regulation of the MRLC kinases is universally present in other cell types, including dedifferentiated vascular smooth muscle cells (SMCs). Here, we demonstrate the absence of the spatial regulation in dedifferentiated SMCs using both cell lines and primary cells. Thus, our work is distinct from previous reports on cells with migratory potential. We also observed that the spatial regulation is partly induced upon fibronectin stimulation and Krüppel-like factor 4 overexpression. To find clues to the mechanism, we reveal how the phosphorylation state of MRLC is determined within dedifferentiated A7r5 SMCs under the enzymatic competition among three major regulators ROCK, MLCK, and MRLC phosphatase (MLCP). We show that ROCK, but not MLCK, predominantly regulates the MRLC phosphorylation in a manner distinct from previous in vitro-based and in silico-based reports. In this ROCK-dominating cellular system, the contractility at physiological conditions was regulated at the level of MRLC diphosphorylation, because its monophosphorylation is already saturated. Thus, the present study provides insights into the molecular basis underlying the absence of spatial MRLC regulation in dedifferentiated SMCs.


Assuntos
Desdiferenciação Celular/fisiologia , Fibronectinas/biossíntese , Fatores de Transcrição Kruppel-Like/biossíntese , Músculo Liso Vascular/metabolismo , Cadeias Leves de Miosina/fisiologia , Animais , Bovinos , Linhagem Celular , Células Cultivadas , Humanos , Fator 4 Semelhante a Kruppel , Quinase de Cadeia Leve de Miosina/metabolismo , Ratos , Quinases Associadas a rho/fisiologia
4.
FASEB J ; 33(3): 3152-3166, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30365366

RESUMO

Myosin light chain 2 ( MYL2) gene encodes the myosin regulatory light chain (RLC) simultaneously in heart ventricles and in slow-twitch skeletal muscle. Using transgenic mice with cardiac-specific expression of the human R58Q-RLC mutant, we sought to determine whether the hypertrophic cardiomyopathy phenotype observed in papillary muscles (PMs) of R58Q mice is also manifested in slow-twitch soleus (SOL) muscles. Skinned SOL muscles and ventricular PMs of R58Q animals exhibited lower contractile force that was not observed in the fast-twitch extensor digitorum longus muscles of R58Q vs. wild-type-RLC mice, but mutant animals did not display gross muscle weakness in vivo. Consistent with SOL muscle abnormalities in R58Q vs. wild-type mice, myosin ATPase staining revealed a decreased proportion of fiber type I/type II only in SOL muscles but not in the extensor digitorum longus muscles. The similarities between SOL muscles and PMs of R58Q mice were further supported by quantitative proteomics. Differential regulation of proteins involved in energy metabolism, cell-cell interactions, and protein-protein signaling was concurrently observed in the hearts and SOL muscles of R58Q mice. In summary, even though R58Q expression was restricted to the heart of mice, functional similarities were clearly observed between the hearts and slow-twitch skeletal muscle, suggesting that MYL2 mutated models of hypertrophic cardiomyopathy may be useful research tools to study the molecular, structural, and energetic mechanisms of cardioskeletal myopathy associated with myosin RLC.-Kazmierczak, K., Liang, J., Yuan, C.-C., Yadav, S., Sitbon, Y. H., Walz, K., Ma, W., Irving, T. C., Cheah, J. X., Gomes, A. V., Szczesna-Cordary, D. Slow-twitch skeletal muscle defects accompany cardiac dysfunction in transgenic mice with a mutation in the myosin regulatory light chain.


Assuntos
Miosinas Cardíacas/genética , Miosinas Cardíacas/fisiologia , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/fisiopatologia , Fibras Musculares de Contração Lenta/fisiologia , Cadeias Leves de Miosina/genética , Cadeias Leves de Miosina/fisiologia , Substituição de Aminoácidos , Animais , Cardiomiopatia Hipertrófica/patologia , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Contração Muscular/genética , Contração Muscular/fisiologia , Fibras Musculares de Contração Lenta/patologia , Mutação de Sentido Incorreto , Contração Miocárdica/genética , Contração Miocárdica/fisiologia , Miocárdio/metabolismo , Miocárdio/patologia , Músculos Papilares/patologia , Músculos Papilares/fisiopatologia , Proteômica
5.
Genesis ; 56(10): e23256, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30307112

RESUMO

MLC-2v is a myosin light chain regulatory protein which is specifically expressed in ventricular cardiomyocytes and slow twitch skeletal muscle cells. MLC-2v plays critical roles in ventricular maturation during heart development. Mice lacking MLC-2v are embryonic lethal due to heart failure associated with abnormal myofibrillar organization of ventricular cardiomyocytes. To study the development of ventricular cardiac muscle and slow twitch skeletal muscle, we generated a new MLC-2v reporter mouse line by knocking-in a tdTomato reporter cassette into 3' UTR of the MLC-2v gene without disrupting the endogenous gene. Our results demonstrated specific MLC-2v-tdTomato knock-in reporter expression in ventricular cardiomyocytes and slow twitch muscle during myogenesis, precisely recapitulating the spatiotemporal expression pattern of endogenous MLC-2v. No tdTomato expression was observed in the atria, fast twitch muscle or other organs throughout development into adulthood. Isolated neonatal and adult ventricular cardiomyocytes uniformly express tdTomato. Taken together, MLC-2v-tdTomato knock-in reporter mouse model described in this article will serve as a valuable tool to study cardiac chamber and skeletal muscle specification during development and regeneration by overcoming the pitfalls of transgenic strategies.


Assuntos
Genes Reporter , Cadeias Leves de Miosina/genética , Animais , Técnicas de Introdução de Genes , Solanum lycopersicum/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Transgênicos , Cadeias Leves de Miosina/fisiologia
6.
Biochem J ; 475(20): 3201-3219, 2018 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-30242065

RESUMO

Deciphering the histone code has illustrated that acetylation or methylation on the same residue can have analogous or opposing roles. However, little is known about the interplay between these post-translational modifications (PTMs) on the same nonhistone residues. We have recently discovered that N-terminal acetyltransferases (NATs) and N-terminal methyltransferases (NRMTs) can have overlapping substrates and identified myosin regulatory light chain 9 (MYL9) as the first confirmed protein to occur in either α-amino-methylated (Nα-methyl) or α-amino-acetylated (Nα-acetyl) states in vivo Here we aim to determine if these PTMs function similarly or create different MYL9 proteoforms with distinct roles. We use enzymatic assays to directly verify MYL9 is a substrate of both NRMT1 and NatA and generate mutants of MYL9 that are exclusive for Nα-acetylation or Nα-methylation. We then employ eukaryotic cell models to probe the regulatory functions of these Nα-PTMs on MYL9. Our results show that, contrary to prevailing dogma, neither of these modifications regulate the stability of MYL9. Rather, exclusive Nα-acetylation promotes cytoplasmic roles of MYL9, while exclusive Nα-methylation promotes the nuclear role of MYL9 as a transcription factor. The increased cytoplasmic activity of Nα-acetylated MYL9 corresponds with increased phosphorylation at serine 19, a key MYL9 activating PTM. Increased nuclear activity of Nα-methylated MYL9 corresponds with increased DNA binding. Nα-methylation also results in a decrease of interactions between the N-terminus of MYL9 and a host of cytoskeletal proteins. These results confirm that Nα-acetylation and Nα-methylation differentially affect MYL9 function by creating distinct proteoforms with different internal PTM patterns and binding properties.


Assuntos
Movimento Celular/fisiologia , Cadeias Leves de Miosina/fisiologia , Acetilação , Animais , Células HCT116 , Células HEK293 , Humanos , Metilação , Camundongos , Células NIH 3T3
7.
Proc Natl Acad Sci U S A ; 114(17): 4483-4488, 2017 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-28396412

RESUMO

Although the neutrophil recruitment cascade during inflammation has been well described, the molecular players that halt neutrophil chemotaxis remain unclear. In this study, we found that lipopolysaccharide (LPS) was a potent stop signal for chemotactic neutrophil migration. Treatment with an antagonist of the ATP receptor (P2X1) in primary human neutrophils or knockout of the P2X1 receptor in neutrophil-like differentiated HL-60 (dHL-60) cells recovered neutrophil chemotaxis. Further observations showed that LPS-induced ATP release through connexin 43 (Cx43) hemichannels was responsible for the activation of the P2X1 receptor and the subsequent calcium influx. Increased intracellular calcium stopped neutrophil chemotaxis by activating myosin light chain (MLC) through the myosin light chain kinase (MLCK)-dependent pathway. Taken together, these data identify a previously unknown function of LPS-induced autocrine ATP signaling in inhibiting neutrophil chemotaxis by enhancing MLC phosphorylation, which provides important evidence that stoppage of neutrophil chemotaxis at infectious foci plays a key role in the defense against invading pathogens.


Assuntos
Trifosfato de Adenosina/fisiologia , Comunicação Autócrina , Quimiotaxia/fisiologia , Endotoxinas/farmacologia , Neutrófilos/fisiologia , Transdução de Sinais/fisiologia , Conexina 43/genética , Conexina 43/metabolismo , Regulação da Expressão Gênica/fisiologia , Células HL-60 , Humanos , Lipopolissacarídeos/farmacologia , Cadeias Leves de Miosina/fisiologia , Fosforilação , Receptores Purinérgicos P2X1
8.
PLoS One ; 12(4): e0174690, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28423017

RESUMO

Myosin motors in cardiac ventriculum convert ATP free energy to the work of moving blood volume under pressure. The actin bound motor cyclically rotates its lever-arm/light-chain complex linking motor generated torque to the myosin filament backbone and translating actin against resisting force. Previous research showed that the unloaded in vitro motor is described with high precision by single molecule mechanical characteristics including unitary step-sizes of approximately 3, 5, and 8 nm and their relative step-frequencies of approximately 13, 50, and 37%. The 3 and 8 nm unitary step-sizes are dependent on myosin essential light chain (ELC) N-terminus actin binding. Step-size and step-frequency quantitation specifies in vitro motor function including duty-ratio, power, and strain sensitivity metrics. In vivo, motors integrated into the muscle sarcomere form the more complex and hierarchically functioning muscle machine. The goal of the research reported here is to measure single myosin step-size and step-frequency in vivo to assess how tissue integration impacts motor function. A photoactivatable GFP tags the ventriculum myosin lever-arm/light-chain complex in the beating heart of a live zebrafish embryo. Detected single GFP emission reports time-resolved myosin lever-arm orientation interpreted as step-size and step-frequency providing single myosin mechanical characteristics over the active cycle. Following step-frequency of cardiac ventriculum myosin transitioning from low to high force in relaxed to auxotonic to isometric contraction phases indicates that the imposition of resisting force during contraction causes the motor to down-shift to the 3 nm step-size accounting for >80% of all the steps in the near-isometric phase. At peak force, the ATP initiated actomyosin dissociation is the predominant strain inhibited transition in the native myosin contraction cycle. The proposed model for motor down-shifting and strain sensing involves ELC N-terminus actin binding. Overall, the approach is a unique bottom-up single molecule mechanical characterization of a hierarchically functional native muscle myosin.


Assuntos
Actinas/química , Actomiosina/química , Miosinas Cardíacas/química , Contração Miocárdica/fisiologia , Miocárdio/metabolismo , Cadeias Leves de Miosina/química , Actinas/genética , Actinas/fisiologia , Actomiosina/genética , Actomiosina/fisiologia , Animais , Fenômenos Biomecânicos , Miosinas Cardíacas/genética , Miosinas Cardíacas/fisiologia , Embrião não Mamífero , Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Contração Isométrica , Miocárdio/ultraestrutura , Cadeias Leves de Miosina/genética , Cadeias Leves de Miosina/fisiologia , Sarcômeros/metabolismo , Sarcômeros/ultraestrutura , Imagem Individual de Molécula , Peixe-Zebra/fisiologia
9.
Dev Biol ; 418(2): 283-96, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27527806

RESUMO

Head development in vertebrates proceeds through a series of elaborate patterning mechanisms and cell-cell interactions involving cephalic neural crest cells (CNCC). These cells undergo extensive migration along stereotypical paths after their separation from the dorsal margins of the neural tube and they give rise to most of the craniofacial skeleton. Here, we report that the silencing of the LKB1 tumor suppressor affects the delamination of pre-migratory CNCC from the neural primordium as well as their polarization and survival, thus resulting in severe facial and brain defects. We further show that LKB1-mediated effects on the development of CNCC involve the sequential activation of the AMP-activated protein kinase (AMPK), the Rho-dependent kinase (ROCK) and the actin-based motor protein myosin II. Collectively, these results establish that the complex morphogenetic processes governing head formation critically depends on the activation of the LKB1 signaling network in CNCC.


Assuntos
Proteínas Aviárias/fisiologia , Crista Neural/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Quinases Ativadas por AMP/fisiologia , Animais , Proteínas Aviárias/antagonistas & inibidores , Proteínas Aviárias/genética , Embrião de Galinha , Anormalidades Craniofaciais/embriologia , Anormalidades Craniofaciais/genética , Regulação da Expressão Gênica no Desenvolvimento , Inativação Gênica , Cabeça/embriologia , Camundongos , Camundongos Knockout , Cadeias Leves de Miosina/fisiologia , Crista Neural/citologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Quinases Associadas a rho/fisiologia
10.
J Physiol ; 594(18): 5237-54, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27291932

RESUMO

KEY POINTS: Regulatory light chain (RLC) phosphorylation has been shown to alter the ability of muscle to produce force and power during shortening and to alter the rate of force redevelopment (ktr ) at submaximal [Ca(2+) ]. Increasing RLC phosphorylation ∼50% from the in vivo level in maximally [Ca(2+) ]-activated cardiac trabecula accelerates ktr . Decreasing RLC phosphorylation to ∼70% of the in vivo control level slows ktr and reduces force generation. ktr is dependent on sarcomere length in the physiological range 1.85-1.94 µm and RLC phosphorylation modulates this response. We demonstrate that Frank-Starling is evident at maximal [Ca(2+) ] activation and therefore does not necessarily require length-dependent change in [Ca(2+) ]-sensitivity of thin filament activation. The stretch response is modulated by changes in RLC phosphorylation, pinpointing RLC phosphorylation as a modulator of the Frank-Starling law in the heart. These data provide an explanation for slowed systolic function in the intact heart in response to RLC phosphorylation reduction. ABSTRACT: Force and power in cardiac muscle have a known dependence on phosphorylation of the myosin-associated regulatory light chain (RLC). We explore the effect of RLC phosphorylation on the ability of cardiac preparations to redevelop force (ktr ) in maximally activating [Ca(2+) ]. Activation was achieved by rapidly increasing the temperature (temperature-jump of 0.5-20ºC) of permeabilized trabeculae over a physiological range of sarcomere lengths (1.85-1.94 µm). The trabeculae were subjected to shortening ramps over a range of velocities and the extent of RLC phosphorylation was varied. The latter was achieved using an RLC-exchange technique, which avoids changes in the phosphorylation level of other proteins. The results show that increasing RLC phosphorylation by 50% accelerates ktr by ∼50%, irrespective of the sarcomere length, whereas decreasing phosphorylation by 30% slows ktr by ∼50%, relative to the ktr obtained for in vivo phosphorylation. Clearly, phosphorylation affects the magnitude of ktr following step shortening or ramp shortening. Using a two-state model, we explore the effect of RLC phosphorylation on the kinetics of force development, which proposes that phosphorylation affects the kinetics of both attachment and detachment of cross-bridges. In summary, RLC phosphorylation affects the rate and extent of force redevelopment. These findings were obtained in maximally activated muscle at saturating [Ca(2+) ] and are not explained by changes in the Ca(2+) -sensitivity of acto-myosin interactions. The length-dependence of the rate of force redevelopment, together with the modulation by the state of RLC phosphorylation, suggests that these effects play a role in the Frank-Starling law of the heart.


Assuntos
Coração/fisiologia , Cadeias Leves de Miosina/fisiologia , Animais , Feminino , Fosforilação , Ratos Sprague-Dawley , Sarcômeros/fisiologia , Função Ventricular
11.
Am J Physiol Cell Physiol ; 310(8): C692-700, 2016 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-26911280

RESUMO

The phosphorylation of the myosin regulatory light chain (RLC) is an important modulator of skeletal muscle performance and plays a key role in posttetanic potentiation and staircase potentiation of twitch contractions. The structural basis for these phenomena within the filament lattice has not been thoroughly investigated. Using a synchrotron radiation source at SPring8, we obtained X-ray diffraction patterns from skinned rabbit psoas muscle fibers before and after phosphorylation of myosin RLC in the presence of myosin light chain kinase, calmodulin, and calcium at a concentration below the threshold for tension development ([Ca(2+)] = 10(-6.8)M). After phosphorylation, the first myosin layer line slightly decreased in intensity at ∼0.05 nm(-1)along the equatorial axis, indicating a partial loss of the helical order of myosin heads along the thick filament. Concomitantly, the (1,1/1,0) intensity ratio of the equatorial reflections increased. These results provide a firm structural basis for the hypothesis that phosphorylation of myosin RLC caused the myosin heads to move away from the thick filaments towards the thin filaments, thereby enhancing the probability of interaction with actin. In contrast, 2,3-butanedione monoxime (BDM), known to inhibit contraction by impeding phosphate release from myosin, had exactly the opposite effects on meridional and equatorial reflections to those of phosphorylation. We hypothesize that these antagonistic effects are due to the acceleration of phosphate release from myosin by phosphorylation and its inhibition by BDM, the consequent shifts in crossbridge equilibria leading to opposite changes in abundance of the myosin-ADP-inorganic phosphate complex state associated with helical order of thick filaments.


Assuntos
Diacetil/análogos & derivados , Contração Muscular/fisiologia , Fibras Musculares Esqueléticas/fisiologia , Fibras Musculares Esqueléticas/ultraestrutura , Cadeias Leves de Miosina/fisiologia , Cadeias Leves de Miosina/ultraestrutura , Animais , Células Cultivadas , Diacetil/farmacologia , Masculino , Contração Muscular/efeitos dos fármacos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Coelhos , Difração de Raios X/métodos
12.
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
13.
Eur J Pharmacol ; 775: 67-77, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26872992

RESUMO

Non-muscle myosin regulatory light chain (nmMLC20) is reported to exert transcriptional function in regulation of gene expression, and NADPH oxidase (NOX)-derived reactive oxygen species contribute to vascular remodeling of pulmonary artery hypertension (PAH). This study aims to determine if nmMLC20 can promote endothelial progenitor cells (EPCs) senescence and dysfunction through up-regulation of NOX in PAH rats. The rats were exposed to10% hypoxia for 3 weeks to establish a PAH model, which showed an increase in right ventricle systolic pressure, right ventricular and pulmonary vascular remodeling, and the accelerated senescence and impaired functions in EPCs, accompanied by an increase in Rho-kinase (ROCK) and NOX activities, p-nmMLC20 level, NOX expression and H2O2 content; these phenomena were reversed by fasudil, a selective inhibitor of ROCK. Next, normal EPCs were cultured under hypoxia to induce senescence in vitro. Consistent with the in vivo findings, hypoxia increased the senescence and dysfunction of EPCs concomitant with an increase in ROCK and NOX activities, p-nmMLC20 level, NOX expression and H2O2 content; these phenomena were reversed by fasudil. Knockdown of nmMLC20 showed similar results to that of fasudil except no effect on ROCK activity. Based on these observations, we conclude that nmMLC20 could promote the senescence and dysfunctions of EPCs in PAH through up-regulation of NOX in a phosphorylation-dependent manner.


Assuntos
Senescência Celular/fisiologia , Células Progenitoras Endoteliais/fisiologia , Hipertensão Pulmonar/metabolismo , Cadeias Leves de Miosina/fisiologia , NADPH Oxidases/metabolismo , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Animais , Hipertensão Pulmonar/etiologia , Hipóxia/complicações , Masculino , Cadeias Leves de Miosina/genética , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Ratos Sprague-Dawley , Regulação para Cima , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/metabolismo
14.
Mol Biol Cell ; 27(7): 1131-42, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26864628

RESUMO

The myoepithelial sheath in the somatic gonad of the nematode Caenorhabditis elegans has nonstriated contractile actomyosin networks that produce highly coordinated contractility for ovulation of mature oocytes. Two myosin heavy chains are expressed in the myoepithelial sheath, which are also expressed in the body-wall striated muscle. The troponin/tropomyosin system is also present and essential for ovulation. Therefore, although the myoepithelial sheath has smooth muscle-like contractile apparatuses, it has a striated muscle-like regulatory mechanism through troponin/tropomyosin. Here we report that the myoepithelial sheath has a distinct myosin population containing nonmuscle myosin II isoforms, which is regulated by phosphorylation and essential for ovulation. MLC-4, a nonmuscle myosin regulatory light chain, localizes to small punctate structures and does not colocalize with large, needle-like myosin filaments containing MYO-3, a striated-muscle myosin isoform. RNA interference of MLC-4, as well as of its upstream regulators, LET-502 (Rho-associated coiled-coil forming kinase) and MEL-11 (a myosin-binding subunit of myosin phosphatase), impairs ovulation. Expression of a phosphomimetic MLC-4 mutant mimicking a constitutively active state also impairs ovulation. A striated-muscle myosin (UNC-54) appears to provide partially compensatory contractility. Thus the results indicate that the two spatially distinct myosin II populations coordinately regulate ovulatory contraction of the myoepithelial sheath.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/fisiologia , Contração Muscular/fisiologia , Cadeias Leves de Miosina/fisiologia , Ovulação/metabolismo , Quinases Associadas a rho/fisiologia , Animais , Caenorhabditis elegans/metabolismo , Feminino , Gônadas/metabolismo , Gônadas/fisiologia
15.
Proc Natl Acad Sci U S A ; 112(33): 10425-30, 2015 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-26240367

RESUMO

To better understand human adaptation to stress, and in particular to hypoxia, we took advantage of one of nature's experiments at high altitude (HA) and studied Ethiopians, a population that is well-adapted to HA hypoxic stress. Using whole-genome sequencing, we discovered that EDNRB (Endothelin receptor type B) is a candidate gene involved in HA adaptation. To test whether EDNRB plays a critical role in hypoxia tolerance and adaptation, we generated EdnrB knockout mice and found that when EdnrB (-/+) heterozygote mice are treated with lower levels of oxygen (O2), they tolerate various levels of hypoxia (even extreme hypoxia, e.g., 5% O2) very well. For example, they maintain ejection fraction, cardiac contractility, and cardiac output in severe hypoxia. Furthermore, O2 delivery to vital organs was significantly higher and blood lactate was lower in EdnrB (-/+) compared with wild type in hypoxia. Tissue hypoxia in brain, heart, and kidney was lower in EdnrB (-/+) mice as well. These data demonstrate that a lower level of EDNRB significantly improves cardiac performance and tissue perfusion under various levels of hypoxia. Transcriptomic profiling of left ventricles revealed three specific genes [natriuretic peptide type A (Nppa), sarcolipin (Sln), and myosin light polypeptide 4 (Myl4)] that were oppositely expressed (q < 0.05) between EdnrB (-/+) and wild type. Functions related to these gene networks were consistent with a better cardiac contractility and performance. We conclude that EDNRB plays a key role in hypoxia tolerance and that a lower level of EDNRB contributes, at least in part, to HA adaptation in humans.


Assuntos
Coração/fisiologia , Hipóxia/patologia , Receptor de Endotelina B/fisiologia , Aclimatação/genética , Altitude , Animais , Fator Natriurético Atrial/fisiologia , Débito Cardíaco/fisiologia , Etiópia , Feminino , Heterozigoto , Humanos , Ácido Láctico/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Musculares/fisiologia , Contração Miocárdica , Cadeias Leves de Miosina/fisiologia , Oxigênio/química , Proteolipídeos/fisiologia , Locos de Características Quantitativas , Receptor de Endotelina B/genética , Análise de Sequência de DNA , Distribuição Tecidual
16.
Basic Res Cardiol ; 110(4): 38, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25982880

RESUMO

Recent studies demonstrated that NADPH oxidase 2 (NOX2) expression in myocardium after ischemia-reperfusion (IR) is significantly upregulated. However, the underlying mechanisms remain unknown. This study aims to determine if nuclear cardiac myosin light chain 2 (MYL2), a well-known regulatory subunit of myosin, functions as a transcription factor to promote NOX2 expression following myocardial IR in a phosphorylation-dependent manner. We examined the phosphorylation status of nuclear MYL2 (p-MYL2) in a rat model of myocardial IR (left main coronary artery subjected to 1 h ligation and 3 h reperfusion) injury, which showed IR injury and upregulated NOX2 expression as expected, accompanied by elevated H2O2 and nuclear p-MYL2 levels; these effects were attenuated by inhibition of myosin light chain kinase (MLCK). Next, we explored the functional relationship of nuclear p-MYL2 with NOX2 expression in H9c2 cell model of hypoxia-reoxygenation (HR) injury. In agreement with our in vivo findings, HR treatment increased apoptosis, NOX2 expression, nuclear p-MYL2 and H2O2 levels, and the increases were ameliorated by inhibition of MLCK or knockdown of MYL2. Finally, molecular biology techniques including co-immunoprecipitation (Co-IP), chromatin immunoprecipitation (ChIP), DNA pull-down and luciferase reporter gene assay were utilized to decipher the molecular mechanisms. We found that nuclear p-MYL2 binds to the consensus sequence AGCTCC in NOX2 gene promoter, interacts with RNA polymerase II and transcription factor IIB to form a transcription preinitiation complex, and thus activates NOX2 gene transcription. Our results demonstrate that nuclear MYL2 plays an important role in IR injury by transcriptionally upregulating NOX2 expression to enhance oxidative stress in a phosphorylation-dependent manner.


Assuntos
Miosinas Cardíacas/fisiologia , Glicoproteínas de Membrana/genética , Miocárdio/metabolismo , Cadeias Leves de Miosina/fisiologia , NADPH Oxidases/genética , Animais , Miosinas Cardíacas/análise , Núcleo Celular/química , Células Cultivadas , Masculino , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Cadeias Leves de Miosina/análise , Quinase de Cadeia Leve de Miosina/antagonistas & inibidores , NADPH Oxidase 2 , Estresse Oxidativo , Fosforilação , Ratos , Ratos Sprague-Dawley
17.
Heart ; 100(24): 1916-23, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25271316

RESUMO

The sarcomere is the principal contractile unit of striated muscle. Mutations in genes encoding sarcomeric proteins are responsible for a range of diseases including hypertrophic, dilated and restrictive cardiomyopathies and ventricular non-compaction. The downstream molecular pathways leading to these heterogeneous phenotypes include changes in acto-myosin cross-bridge kinetics, altered mechanosensation, disturbed calcium sensitivity, de-regulated signalling pathways, inefficient energetics, myocardial ischaemia and fibrosis. The elucidation of the genetic causes of cardiomyopathy has helped in understanding the structure and function of the sarcomere and a more detailed knowledge of the sarcomere and its associated proteins has suggested additional gene candidates. The new hope is that these advances will stimulate the discovery of disease-modifying drugs.


Assuntos
Cardiomiopatias/etiologia , Sarcômeros/fisiologia , Cálcio/fisiologia , Cardiomiopatias/fisiopatologia , Cardiomiopatias/terapia , Proteínas de Transporte/fisiologia , Comunicação Celular/fisiologia , Terapia Genética/métodos , Homeostase/fisiologia , Humanos , Mutação/genética , Contração Miocárdica/fisiologia , Miofibroblastos/fisiologia , Cadeias Pesadas de Miosina/fisiologia , Cadeias Leves de Miosina/fisiologia , Sarcômeros/química , Sarcômeros/genética , Transdução de Sinais/fisiologia
18.
J Biol Chem ; 289(44): 30832-30841, 2014 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-25231988

RESUMO

Many diverse myosin classes can be expressed using the baculovirus/Sf9 insect cell expression system, whereas others have been recalcitrant. We hypothesized that most myosins utilize Sf9 cell chaperones, but others require an organism-specific co-chaperone. TgMyoA, a class XIVa myosin from the parasite Toxoplasma gondii, is required for the parasite to efficiently move and invade host cells. The T. gondii genome contains one UCS family myosin co-chaperone (TgUNC). TgMyoA expressed in Sf9 cells was soluble and functional only if the heavy and light chain(s) were co-expressed with TgUNC. The tetratricopeptide repeat domain of TgUNC was not essential to obtain functional myosin, implying that there are other mechanisms to recruit Hsp90. Purified TgMyoA heavy chain complexed with its regulatory light chain (TgMLC1) moved actin in a motility assay at a speed of ∼1.5 µm/s. When a putative essential light chain (TgELC1) was also bound, TgMyoA moved actin at more than twice that speed (∼3.4 µm/s). This result implies that two light chains bind to and stabilize the lever arm, the domain that amplifies small motions at the active site into the larger motions that propel actin at fast speeds. Our results show that the TgMyoA domain structure is more similar to other myosins than previously appreciated and provide a molecular explanation for how it moves actin at fast speeds. The ability to express milligram quantities of a class XIV myosin in a heterologous system paves the way for detailed structure-function analysis of TgMyoA and identification of small molecule inhibitors.


Assuntos
Chaperonas Moleculares/biossíntese , Cadeias Pesadas de Miosina/química , Cadeias Leves de Miosina/fisiologia , Proteínas de Protozoários/química , Toxoplasma/metabolismo , Actinas/química , Animais , Transporte Biológico , Cálcio/química , Chaperonas Moleculares/química , Cadeias Pesadas de Miosina/biossíntese , Cadeias Leves de Miosina/química , Ligação Proteica , Proteínas de Protozoários/fisiologia , Células Sf9 , Solubilidade , Spodoptera
19.
Eur J Pharmacol ; 720(1-3): 355-62, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24120660

RESUMO

Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid that contracts most smooth muscles. Although S1P has been shown to contract bladder smooth muscle, the mechanism(s) by which S1P initiates contraction has not been extensively investigated. The goal of this study was to determine if S1P-induced force generation and myosin light chain (MLC) phosphorylation are dependent on calcium sensitization pathways mediated by protein kinase C (PKC) and Rho kinase (ROCK) and which S1P receptor is important in this response. Bladder smooth muscle strips from rabbit and rat were mounted for isometric force recording and contracted in response to carbachol or S1P in the presence and absence of an inhibitor of PKC (3 µM Bisindolylmaleimide-1) or ROCK (1 µM H-1172). 10 µM S1P produced approximately 40% of the force generated in response to 110 mM KCl in rabbit bladder smooth muscle. S1P, up to 100 µM, did not produce a response in rat bladder smooth muscle, any response evoked was due to solvent (NaOH). S1P-dependent force development was associated with a concomitant increase in Ser(19), but not dual Thr(18)/Ser(19) MLC phosphorylation. Inhibition of PKC decreased force development, whereas inhibition of ROCK abolished S1P-induced force. An inhibitor of the S1P2 receptor, JTE-013, relaxed a S1P-induced contraction; whereas, an agonist with low affinity to the S1P2 receptor, dihydro-S1P, did not elicit a contraction. Our results suggest that S1P contracts rabbit, but not rat, bladder smooth muscle via the S1P2 receptor and is dependent on MLC phosphorylation and myofilament calcium sensitization primarily in response to ROCK activation.


Assuntos
Contração Isométrica/fisiologia , Lisofosfolipídeos/fisiologia , Músculo Liso/fisiologia , Esfingosina/análogos & derivados , Bexiga Urinária/fisiologia , Animais , Técnicas In Vitro , Masculino , Cadeias Leves de Miosina/fisiologia , Fosforilação , Proteína Quinase C/fisiologia , Coelhos , Ratos , Ratos Zucker , Esfingosina/fisiologia , Quinases Associadas a rho/fisiologia
20.
J Muscle Res Cell Motil ; 34(5-6): 317-32, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24162313

RESUMO

The contractile performance of mammalian fast twitch skeletal muscle is history dependent. The effect of previous or ongoing contractile activity to potentiate force, i.e. increase isometric twitch force, is a fundamental property of fast skeletal muscle. The precise manifestation of force potentiation is dependent upon a variety of factors with two general types being identified; staircase potentiation referring to the progressive increase in isometric twitch force observed during low frequency stimulation while posttetanic potentiation refers to the step-like increase in isometric twitch force observed following a brief higher frequency (i.e. tetanic) stimulation. Classic studies established that the magnitude and duration of potentiation depends on a number of factors including muscle fiber type, species, temperature, sarcomere length and stimulation paradigm. In addition to isometric twitch force, more recent work has shown that potentiation also influences dynamic (i.e. concentric and/or isotonic) force, work and power at a range of stimulus frequencies in situ or in vitro, an effect that may translate to enhanced physiological function in vivo. Early studies performed on both intact and permeabilized models established that the primary mechanism for this modulation of performance was phosphorylation of myosin, a modification that increased the Ca(2+) sensitivity of contraction. More recent work from a variety of muscle models indicates, however, the presence of a secondary mechanism for potentiation that may involve altered Ca(2+) handling. The primary purpose of this review is to highlight these recent findings relative to the physiological utility of force potentiation in vivo.


Assuntos
Contração Isométrica/fisiologia , Músculo Esquelético/fisiologia , Cadeias Leves de Miosina/fisiologia , Animais , Humanos , Modelos Animais , Músculo Esquelético/metabolismo , Cadeias Leves de Miosina/metabolismo , Fosforilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...