Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 549
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biol Pharm Bull ; 47(7): 1368-1375, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39085076

RESUMEN

We previously reported that the sustained component of contraction induced by depolarizing stimulation by high K+ concentration in rat caudal arterial smooth muscle involves a Ca2+-induced Ca2+ sensitization mechanism whereby Ca2+ entry through voltage-gated Ca2+ channels activates proline-rich tyrosine kinase 2 (Pyk2), leading to activation of RhoA/Rho-associated kinase (ROCK). In the present study, we investigated a potential role for Pyk2-mediated RhoA/ROCK activation in contraction mediated by elevation of cytosolic free Ca2+ concentration ([Ca2+]i) induced by a Ca2+ ionophore, ionomycin, rather than by depolarizing stimulation. Ionomycin (60 µM) induced slow and sustained contraction of rat caudal arterial smooth muscle due to influx of Ca2+. Pre-treatment with a myosin light chain kinase (MLCK) inhibitor, ML-9 (30 µM), inhibited both the early phase (4 min) and the sustained phase (30 min) of ionomycin-induced contraction. On the other hand, a ROCK inhibitor, HA-1077 (3 µM), and Pyk2 inhibitors, sodium salicylate (10 mM) and PF-431396 (3 µM), suppressed only the sustained phase of ionomycin-induced contraction. A calmodulin (CaM) inhibitor, W-7 (150 µM), but not W-5 (150 µM), suppressed the early phase of contraction. Early or sustained increase of ionomycin-induced 20 kDa light chain of myosin (LC20) phosphorylation was inhibited by each inhibitor in a manner similar to the attenuation of contraction. These results indicate that the early phase of ionomycin-induced contraction is mediated by MLCK activation by [Ca2+]i elevation, whereas the sustained phase of ionomycin-induced contraction involves RhoA/ROCK activation and inhibition of myosin light chain phosphatase (MLCP) through CaM-independent Pyk2 activation by [Ca2+]i elevation.


Asunto(s)
Calcio , Ionomicina , Contracción Muscular , Quinasas Asociadas a rho , Animales , Ionomicina/farmacología , Masculino , Contracción Muscular/efectos de los fármacos , Calcio/metabolismo , Quinasas Asociadas a rho/metabolismo , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Ratas , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiología , Músculo Liso Vascular/metabolismo , Quinasa 2 de Adhesión Focal/metabolismo , Ionóforos de Calcio/farmacología , Proteína de Unión al GTP rhoA/metabolismo , Ratas Sprague-Dawley , Ratas Wistar , Calmodulina/metabolismo
2.
Mol Hum Reprod ; 26(4): 228-239, 2020 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-32119740

RESUMEN

Zinc dynamics are essential for oocyte meiotic maturation, egg activation, and preimplantation embryo development. During fertilisation and egg activation, the egg releases billions of zinc atoms (Zn2+) in an exocytotic event termed the 'zinc spark'. We hypothesised that this zinc transport and exocytosis is dependent upon the intracellular trafficking of cortical granules (CG) which requires myosin-actin-dependent motors. Treatment of mature mouse and human eggs with ML-7, a myosin light chain kinase inhibitor (MLCK), resulted in an 80% reduction in zinc spark intensity compared to untreated controls when activated with ionomycin. Moreover, CG migration towards the plasma membrane was significantly decreased in ML-7-treated eggs compared with controls when activated parthenogenetically with ionomycin. In sperm-induced fertilisation via intracytoplasmic sperm injection (ICSI), ML-7-treated mouse eggs exhibited decreased labile zinc intensity and cortical CG staining. Collectively, these data demonstrate that ML-7 treatment impairs zinc release from both murine and human eggs after activation, demonstrating that zinc exocytosis requires myosin light chain kinase activity. Further, these results provide additional support that zinc is likely stored and released from CGs. These data underscore the importance of intracellular zinc trafficking as a crucial component of egg maturation necessary for egg activation and early embryo development.


Asunto(s)
Exocitosis , Quinasa de Cadena Ligera de Miosina/metabolismo , Óvulo/metabolismo , Adulto , Animales , Azepinas , Femenino , Humanos , Técnicas In Vitro , Masculino , Ratones , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Naftalenos , Oogénesis , Óvulo/citología , Inyecciones de Esperma Intracitoplasmáticas
3.
J Cell Mol Med ; 23(5): 3696-3710, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30907506

RESUMEN

To identify the interaction between known regulators of atherosclerosis, microRNA-92a (miR-92a), Rho-associated coiled-coil-forming kinase (ROCK) and myosin light chain kinase (MLCK), we examined their expressions during proliferation and migration of platelet-derived growth factor-BB (PDGF-BB)-regulated vascular smooth muscle cells (VSMCs), both in vivo and in vitro. During the formation of atherosclerosis plaque in mice, a parallel increase in expression levels of MLCK and miR-92a was observed while miR-92a expression was reduced in ML-7 (an inhibitor of MLCK) treated mice and in MLCK-deficient VSMCs. In vitro results indicated that both MLCK and miR-92a shared the same signalling pathway. Transfection of miR-92a mimic partially restored the effect of MLCK's deficiency and antagonized the effect of Y27632 (an inhibitor of ROCK) on the down-regulation of VSMCs activities. ML-7 increased the expression of Kruppel-like factor 4 (KLF4, a target of miR-92a), and siRNA-KLF4 increased VSMCs' activity level. Consistently, inhibition of either MLCK or ROCK enhanced the KLF4 expression. Moreover, we observed that ROCK/MLCK up-regulated miR-92a expression in VSMCs through signal transducer and activator of transcription 3 (STAT3) activation. In conclusion, the activation of ROCK/STAT3 and/or MLCK/STAT3 may up-regulate miR-92a expression, which subsequently inhibits KLF4 expression and promotes PDGF-BB-mediated proliferation and migration of VSMCs. This new downstream node in the ROCK/MLCK signalling pathway may offer a potential intervention target for treatment of atherosclerosis.


Asunto(s)
Movimiento Celular/genética , Proliferación Celular/genética , MicroARNs/genética , Miocitos del Músculo Liso/metabolismo , Quinasa de Cadena Ligera de Miosina/genética , Quinasas Asociadas a rho/genética , Animales , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Azepinas/farmacología , Becaplermina/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Factor 4 Similar a Kruppel , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/efectos de los fármacos , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/metabolismo , Naftalenos/farmacología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Quinasas Asociadas a rho/metabolismo
4.
Mediators Inflamm ; 2018: 6434910, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30210262

RESUMEN

Intestinal epithelial barrier damage disrupts immune homeostasis and leads to many intestinal disorders. Lactobacillus reuteri strains have probiotic functions in their modulation of the microbiota and immune system in intestines. In this study, the effects of L. reuteri LR1, a new strain isolated from the feces of weaning piglets, on intestinal epithelial barrier damage in IPEC-1 cells caused by challenge with enterotoxigenic Escherichia coli (ETEC) K88 were examined. It was found that L. reuteri LR1, in large part, offset the ETEC K88-induced increase in permeability of IPEC-1 cell monolayers and decreased the adhesion and invasion of the coliform in IPEC-1 cells. In addition, L. reuteri LR1 increased transcript abundance and protein contents of tight junction (TJ) proteins zonula occluden-1 (ZO-1) and occludin in ETEC K88-infected IPEC-1 cells, whereas it had no effects on claudin-1 and F-actin expression. Using colloidal gold immunoelectron microscopy, these effects of L. reuteri LR1 on ZO-1 and occludin content in IPEC-1 cells were confirmed. By using ML-7, a selective inhibitor of myosin light-chain kinase (MLCK), the beneficial effect of L. reuteri LR1 on contents of ZO-1 and occludin was shown to be dependent on the MLCK pathway. In conclusion, L. reuteri LR1 had beneficial effects on epithelial barrier function consistent with increasing ZO-1 and occludin expression via a MLCK-dependent manner in IPEC-1 cells during challenge with ETEC K88.


Asunto(s)
Escherichia coli Enterotoxigénica/patogenicidad , Limosilactobacillus reuteri/fisiología , Quinasa de Cadena Ligera de Miosina/metabolismo , Proteínas de Uniones Estrechas/metabolismo , Animales , Azepinas/farmacología , Línea Celular , Microscopía Inmunoelectrónica , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Naftalenos/farmacología , Ocludina/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Porcinos , Proteína de la Zonula Occludens-1/metabolismo
5.
Am J Physiol Endocrinol Metab ; 310(9): E724-33, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26956186

RESUMEN

Impairment of skeletal muscle function has been associated with changes in ovarian hormones, especially estradiol. To elucidate mechanisms of estradiol on skeletal muscle strength, the hormone's effects on phosphorylation of the myosin regulatory light chain (pRLC) and muscle contractility were investigated, hypothesizing an estradiol-specific beneficial impact. In a skeletal muscle cell line, C2C12, pRLC was increased by 17ß-estradiol (E2) in a concentration-dependent manner. In skeletal muscles of C57BL/6 mice that were E2 deficient via ovariectomy (OVX), pRLC was lower than that from ovary-intact, sham-operated mice (Sham). The reduced pRLC in OVX muscle was reversed by in vivo E2 treatment. Posttetanic potentiation (PTP) of muscle from OVX mice was low compared with that from Sham mice, and this decrement was reversed by acute E2 treatment, demonstrating physiological consequence. Western blot of those muscles revealed that low PTP corresponded with low pRLC and higher PTP with greater pRLC. We aimed to elucidate signaling pathways affecting E2-mediated pRLC using a kinase inhibitor library and C2C12 cells as well as a specific myosin light chain kinase inhibitor in muscles. PI3K/Akt, MAPK, and CamKII were identified as candidate kinases sensitive to E2 in terms of phosphorylating RLC. Applying siRNA strategy in C2C12 cells, pRLC triggered by E2 was found to be mediated by estrogen receptor-ß and the G protein-coupled estrogen receptor. Together, these results provide evidence that E2 modulates myosin pRLC in skeletal muscle and is one mechanism by which this hormone can affect muscle contractility in females.


Asunto(s)
Estradiol/farmacología , Estrógenos/farmacología , Contracción Muscular/efectos de los fármacos , Fibras Musculares Esqueléticas/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Cadenas Ligeras de Miosina/efectos de los fármacos , Animales , Western Blotting , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Línea Celular , Estradiol/metabolismo , Receptor beta de Estrógeno/genética , Femenino , Técnicas de Silenciamiento del Gen , Ratones , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/metabolismo , Ovariectomía , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , ARN Interferente Pequeño , Receptores de Estrógenos/genética , Receptores Acoplados a Proteínas G/genética , Transducción de Señal
6.
Nutr Cancer ; 68(7): 1225-33, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27564600

RESUMEN

All-trans-retinoic acid (ATRA) inhibits the invasive and metastatic potentials of various cancer cells. However, the underlying mechanism is unclear. Here, we demonstrate that ATRA inhibited colorectal cancer cells RKO (human colon adenocarcinoma cell) migration by downregulating cell movement and increasing cell adhesion. ATRA inhibited the expression and activation of myosin light chain kinase (MLCK) in RKO cells, while the expression level of MLC phosphatase (MLCP) had no change in RKO cells treated with or without ATRA. The expression and activity of MLC was also inhibited in RKO cells exposed to ATRA. Intriguingly, ATRA increased the expression of occludin messenger RNA (mRNA) and protein and its localization on cell membrane. However, ATRA did not change the expression of zonula occludens 1 (ZO-1), but increased the accumulation of ZO-1 on RKO cells membrane. ML-7, an inhibitor of MLCK, significantly inhibited RKO cell migration. Furthermore, knockdown of endogenous MLCK expression inhibited RKO migration. Mechanistically, we showed that MAPK-specific inhibitor PD98059 enhanced the inhibitory effect of ATRA on RKO migration. In contrast, phorbol 12-myristate 13-acetate (PMA) attenuated the effects of ATRA in RKO cells. Moreover, knocking down endogenous extracellular signal-regulated kinase (ERK) expression inhibited MLCK expression in the RKO cells. In conclusion, ATRA inhibits RKO migration by reducing MLCK expression via extracellular signal-regulated kinase 1/Mitogen-activated protein kinase (ERK1/MAPK) signaling pathway.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Regulación hacia Abajo/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Tretinoina/farmacología , Carcinógenos/farmacología , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Membrana Celular/patología , Movimiento Celular/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Activación Enzimática/efectos de los fármacos , Humanos , Cinética , Quinasa de Cadena Ligera de Miosina/genética , Quinasa de Cadena Ligera de Miosina/metabolismo , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Ocludina/agonistas , Ocludina/antagonistas & inhibidores , Ocludina/genética , Ocludina/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Uniones Estrechas/patología , Proteína de la Zonula Occludens-1/agonistas , Proteína de la Zonula Occludens-1/antagonistas & inhibidores , Proteína de la Zonula Occludens-1/genética , Proteína de la Zonula Occludens-1/metabolismo
7.
J Pept Sci ; 22(11-12): 673-681, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27699916

RESUMEN

Myosin light chain kinase (MLCK) is a key regulator of various forms of cell motility including smooth muscle contraction, cell migration, cytokinesis, receptor capping, secretion, etc. Inhibition of MLCK activity in endothelial and epithelial monolayers using cell-permeant peptide Arg-Lys-Lys-Tyr-Lys-Tyr-Arg-Arg-Lys (PIK, Peptide Inhibitor of Kinase) allows protecting the barrier capacity, suggesting a potential medical use of PIK. However, low stability of L-PIK in a biological milieu prompts for development of more stable L-PIK analogues for use as experimental tools in basic and drug-oriented biomedical research. Previously, we designed PIK1, H-(Nα Me)Arg-Lys-Lys-Tyr-Lys-Tyr-Arg-Arg-Lys-NH2 , that was 2.5-fold more resistant to peptidases in human plasma in vitro than L-PIK and equal to it as MLCK inhibitor. In order to further enhance proteolytic stability of PIK inhibitor, we designed the set of six site-protected peptides based on L-PIK and PIK1 degradation patterns in human plasma as revealed by 1 H-NMR analysis. Implemented modifications increased half-live of the PIK-related peptides in plasma about 10-fold, and these compounds retained 25-100% of L-PIK inhibitory activity toward MLCK in vitro. Based on stability and functional activity ranking, PIK2, H-(Nα Me)Arg-Lys-Lys-Tyr-Lys-Tyr-Arg-D-Arg-Lys-NH2 , was identified as the most stable and effective L-PIK analogue. PIK2 was able to decrease myosin light chain phosphorylation in endothelial cells stimulated with thrombin, and this effect correlated with the inhibition by PIK2 of thrombin-induced endothelial hyperpermeability in vitro. Therefore, PIK2 could be used as novel alternative to other cell-permeant inhibitors of MLCK in cell culture-based and in vivo studies where MLCK catalytic activity inhibition is required. Copyright © 2016 European Peptide Society and John Wiley & Sons, Ltd.


Asunto(s)
Proteínas Aviares/antagonistas & inhibidores , Péptidos de Penetración Celular/síntesis química , Células Endoteliales/efectos de los fármacos , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/síntesis química , Secuencia de Aminoácidos , Animales , Proteínas Aviares/química , Proteínas Aviares/aislamiento & purificación , Química Encefálica , Bovinos , Línea Celular , Péptidos de Penetración Celular/sangre , Péptidos de Penetración Celular/farmacología , Células Endoteliales/citología , Células Endoteliales/enzimología , Molleja de las Aves/química , Semivida , Humanos , Quinasa de Cadena Ligera de Miosina/química , Quinasa de Cadena Ligera de Miosina/aislamiento & purificación , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/sangre , Inhibidores de Proteínas Quinasas/farmacología , Estabilidad Proteica , Proteolisis , Técnicas de Síntesis en Fase Sólida/métodos , Trombina/antagonistas & inhibidores , Trombina/farmacología , Pavos
8.
Nature ; 467(7317): 859-62, 2010 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-20944748

RESUMEN

Herpes simplex virus-1 (HSV-1), the prototype of the α-herpesvirus family, causes life-long infections in humans. Although generally associated with various mucocutaneous diseases, HSV-1 is also involved in lethal encephalitis. HSV-1 entry into host cells requires cellular receptors for both envelope glycoproteins B (gB) and D (gD). However, the gB receptors responsible for its broad host range in vitro and infection of critical targets in vivo remain unknown. Here we show that non-muscle myosin heavy chain IIA (NMHC-IIA), a subunit of non-muscle myosin IIA (NM-IIA), functions as an HSV-1 entry receptor by interacting with gB. A cell line that is relatively resistant to HSV-1 infection became highly susceptible to infection by this virus when NMHC-IIA was overexpressed. Antibody to NMHC-IIA blocked HSV-1 infection in naturally permissive target cells. Furthermore, knockdown of NMHC-IIA in the permissive cells inhibited HSV-1 infection as well as cell-cell fusion when gB, gD, gH and gL were coexpressed. Cell-surface expression of NMHC-IIA was markedly and rapidly induced during the initiation of HSV-1 entry. A specific inhibitor of myosin light chain kinase, which regulates NM-IIA by phosphorylation, reduced the redistribution of NMHC-IIA as well as HSV-1 infection in cell culture and in a murine model for herpes stromal keratitis. NMHC-IIA is ubiquitously expressed in various human tissues and cell types and, therefore, is implicated as a functional gB receptor that mediates broad HSV-1 infectivity both in vitro and in vivo. The identification of NMHC-IIA as an HSV-1 entry receptor and the involvement of NM-IIA regulation in HSV-1 infection provide an insight into HSV-1 entry and identify new targets for antiviral drug development.


Asunto(s)
Herpesvirus Humano 1/fisiología , Miosina Tipo IIA no Muscular/metabolismo , Receptores Virales/metabolismo , Adsorción , Animales , Azepinas/farmacología , Células CHO , Fusión Celular , Chlorocebus aethiops , Cricetinae , Cricetulus , Femenino , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HL-60 , Herpes Simple/virología , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 1/metabolismo , Humanos , Ratones , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Naftalenos/farmacología , Miosina Tipo IIA no Muscular/deficiencia , Miosina Tipo IIA no Muscular/genética , Temperatura , Regulación hacia Arriba , Células Vero , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus/efectos de los fármacos
9.
Acta Pharmacol Sin ; 37(8): 1091-100, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27345626

RESUMEN

AIM: Postoperative ileus (POI) is a postoperative dysmotility disorder of gastrointestinal tract, which remains one of the most perplexing problems in medicine. In the present study we investigated the effects of hesperidin, a major flavonoid in sweet oranges and lemons, on POI in rats. METHODS: SD rats were administered hesperidin (5, 20, and 80 mg·kg(-1)·d(-1), ig) for 3 consecutive days. POI operation (gently manipulating the cecum for 1 min) was performed on d 2. The gastrointestinal motility and isolated intestinal contraction were examined 1 d after the operation. Then the myosin phosphorylation and inflammatory responses in cecum tissue were assessed. Smooth muscle cells were isolated from rat small intestine for in vitro experiments. RESULTS: The gastric emptying and intestinal transit were significantly decreased in POI rats, which were reversed by administration of hesperidin. In ileum and cecum preparations of POI rats in vitro, hesperidin (2.5-160 µmol/L) dose-dependently increased the spontaneous contraction amplitudes without affecting the contractile frequency, which was blocked by the myosin light chain kinase (MLCK) inhibitor ML-7 or verapamil, but not by TTX. Furthermore, administration of hesperidin increased the phosphorylation of MLC20 in the cecum tissue of POI rats. Moreover, administration of hesperidin reversed the increased levels of inflammatory cytokines, iNOS and COX-2 in cecum tissue of POI rats. In freshly isolated intestinal smooth muscle cells, hesperidin (5-80 µmol/L) dose-dependently increased the intracellular Ca(2+) concentration as well as the phosphorylation of MLC20, which was abrogated by ML-7 or siRNA that knocked down MLCK. CONCLUSION: Oral administration of hesperidin effectively alleviates rat POI through inhibition of inflammatory responses and stimulation of Ca(2+)-dependent MLC phosphorylation.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Hesperidina/farmacología , Ileus/tratamiento farmacológico , Inflamación/prevención & control , Miosinas/metabolismo , Fosforilación/efectos de los fármacos , Animales , Antiinflamatorios no Esteroideos/antagonistas & inhibidores , Antiinflamatorios no Esteroideos/uso terapéutico , Azepinas/farmacología , Calcio/metabolismo , Ciego/metabolismo , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Vaciamiento Gástrico/efectos de los fármacos , Motilidad Gastrointestinal/efectos de los fármacos , Hesperidina/antagonistas & inhibidores , Hesperidina/uso terapéutico , Intestino Delgado/fisiología , Masculino , Contracción Muscular/efectos de los fármacos , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Naftalenos/farmacología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Complicaciones Posoperatorias/tratamiento farmacológico , ARN Interferente Pequeño/farmacología , Ratas , Verapamilo/farmacología
10.
Dig Dis Sci ; 61(8): 2252-2261, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27074920

RESUMEN

BACKGROUND: IL-1ß is a cytokine involved in mediating epithelial barrier dysfunction in the gut. It is known that IL-1ß mediates activation of non-muscle myosin light chain kinase in epithelial cells, but the precise mechanism by which epithelial barrier dysfunction is induced by IL-1ß is not understood. METHODS AND RESULTS: Using a Caco2 cell model, we show that the expression of the tight junction protein, claudin-3, is transcriptionally downregulated by IL-1ß treatment. In addition, after assessing protein and mRNA expression, and protein localization, we show that inhibition of nmMLCK rescues IL-1ß-mediated decrease in claudin-3 expression as well as junction protein redistribution. Using chromatin immunoprecipitation assays, we also show that ß-catenin targeting of the claudin-3 promoter occurs as a consequence of IL-1ß-mediated epithelial barrier dysfunction, and inhibition of nmMLCK interferes with this interaction. CONCLUSIONS: Taken together, these data represent the first line of evidence demonstrating nmMLCK regulation of claudin-3 expression in response to IL-1ß-treated epithelial cells.


Asunto(s)
Claudina-3/efectos de los fármacos , Interleucina-1beta/farmacología , Mucosa Intestinal/diagnóstico por imagen , Permeabilidad/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , Uniones Estrechas/efectos de los fármacos , beta Catenina/efectos de los fármacos , Azepinas/farmacología , Células CACO-2 , Inmunoprecipitación de Cromatina , Claudina-3/genética , Claudina-3/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Inmunohistoquímica , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Microscopía Confocal , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/metabolismo , Naftalenos/farmacología , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Uniones Estrechas/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
11.
Proc Natl Acad Sci U S A ; 110(43): 17253-8, 2013 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-24101477

RESUMEN

Phenotypic polarization of macrophages is regulated by a milieu of cues in the local tissue microenvironment. Although much is known about how soluble factors influence macrophage polarization, relatively little is known about how physical cues present in the extracellular environment might modulate proinflammatory (M1) vs. prohealing (M2) activation. Specifically, the role of cell shape has not been explored, even though it has been observed that macrophages adopt different geometries in vivo. We and others observed that macrophages polarized toward different phenotypes in vitro exhibit dramatic changes in cell shape: M2 cells exhibit an elongated shape compared with M1 cells. Using a micropatterning approach to control macrophage cell shape directly, we demonstrate here that elongation itself, without exogenous cytokines, leads to the expression of M2 phenotype markers and reduces the secretion of inflammatory cytokines. Moreover, elongation enhances the effects of M2-inducing cytokines IL-4 and IL-13 and protects cells from M1-inducing stimuli LPS and IFN-γ. In addition shape- but not cytokine-induced polarization is abrogated when actin and actin/myosin contractility are inhibited by pharmacological agents, suggesting a role for the cytoskeleton in the control of macrophage polarization by cell geometry. Our studies demonstrate that alterations in cell shape associated with changes in ECM architecture may provide integral cues to modulate macrophage phenotype polarization.


Asunto(s)
Biomarcadores/análisis , Forma de la Célula/inmunología , Citocinas/inmunología , Mediadores de Inflamación/inmunología , Macrófagos/inmunología , Amidas/farmacología , Animales , Arginasa/inmunología , Arginasa/metabolismo , Azepinas/farmacología , Biomarcadores/metabolismo , Polaridad Celular/efectos de los fármacos , Polaridad Celular/inmunología , Forma de la Célula/efectos de los fármacos , Células Cultivadas , Citocalasina D/farmacología , Citocinas/metabolismo , Doxorrubicina/análogos & derivados , Doxorrubicina/inmunología , Doxorrubicina/metabolismo , Femenino , Citometría de Flujo , Inmunofenotipificación/métodos , Mediadores de Inflamación/metabolismo , Interferón gamma/farmacología , Interleucina-13/farmacología , Interleucina-4/farmacología , Lectinas Tipo C/inmunología , Lectinas Tipo C/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/metabolismo , Receptor de Manosa , Lectinas de Unión a Manosa/inmunología , Lectinas de Unión a Manosa/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/metabolismo , Piridinas/farmacología , Receptores de Superficie Celular/inmunología , Receptores de Superficie Celular/metabolismo
12.
Int J Mol Sci ; 17(10)2016 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-27735862

RESUMEN

As a physiological small molecular product from the microbial fermentation of dietary fibers, butyrate plays an important role in maintaining intestinal health. Our previous works have proved that the effect of sodium butyrate (NaB) on the intestinal barrier function is mediated by activation of AMP-activated protein kinase (AMPK). However, the detailed pathway involved remains unknown. Using the calcium switch assay in the Caco-2 cell monolayer model, we found here that NaB activated AMPK mainly by increasing the calcium level, but not the ATP concentration, via promoting store-operated calcium entry (SOCE). Upon the activation of AMPK, NaB promoted the reassembly of tight junctions (TJs) based on reducing the phosphorylation of myosin II regulatory light chain (MLC2) at Ser19 and increasing phosphorylation of protein kinase C ß2 (PKCß2) at Ser660. Inhibiting (protein kinase C ß) PKCß blocked the reassembly of TJs induced by NaB in the barrier monolayer model. These results indicated that NaB could activate the calcium/calmodulin-dependent protein kinase kinase ß (CaMKKß) pathway to mediate AMPK phosphorylating, which then inhibited the phosphorylation of MLC2 and promoted the phosphorylation of PKCß2, respectively, so that the downstream molecules of AMPK coordinately contributed to the reassembly of TJs in the Caco-2 barrier model. These results suggested a potential mechanism of butyrate for intestine homeostasis and protection.


Asunto(s)
Ácido Butírico/farmacología , Miosinas Cardíacas/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Proteína Quinasa C beta/metabolismo , Uniones Estrechas/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/metabolismo , Western Blotting , Células CACO-2 , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Miosinas Cardíacas/antagonistas & inhibidores , Humanos , Inmunoprecipitación , Cadenas Ligeras de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Fosforilación/efectos de los fármacos , Uniones Estrechas/metabolismo
13.
Biophys J ; 109(4): 670-86, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26287620

RESUMEN

The nucleus has a smooth, regular appearance in normal cells, and its shape is greatly altered in human pathologies. Yet, how the cell establishes nuclear shape is not well understood. We imaged the dynamics of nuclear shaping in NIH3T3 fibroblasts. Nuclei translated toward the substratum and began flattening during the early stages of cell spreading. Initially, nuclear height and width correlated with the degree of cell spreading, but over time, reached steady-state values even as the cell continued to spread. Actomyosin activity, actomyosin bundles, microtubules, and intermediate filaments, as well as the LINC complex, were all dispensable for nuclear flattening as long as the cell could spread. Inhibition of actin polymerization as well as myosin light chain kinase with the drug ML7 limited both the initial spreading of cells and flattening of nuclei, and for well-spread cells, inhibition of myosin-II ATPase with the drug blebbistatin decreased cell spreading with associated nuclear rounding. Together, these results show that cell spreading is necessary and sufficient to drive nuclear flattening under a wide range of conditions, including in the presence or absence of myosin activity. To explain this observation, we propose a computational model for nuclear and cell mechanics that shows how frictional transmission of stress from the moving cell boundaries to the nuclear surface shapes the nucleus during early cell spreading. Our results point to a surprisingly simple mechanical system in cells for establishing nuclear shapes.


Asunto(s)
Movimiento Celular/fisiología , Forma del Núcleo Celular/fisiología , Núcleo Celular/fisiología , Fibroblastos/citología , Fibroblastos/fisiología , Células 3T3 , Actinas/metabolismo , Actomiosina/metabolismo , Animales , Azepinas/farmacología , Movimiento Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Forma del Núcleo Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Fibroblastos/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Ratones , Microtúbulos/metabolismo , Modelos Biológicos , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/metabolismo , Miosinas/antagonistas & inhibidores , Miosinas/metabolismo , Naftalenos/farmacología
14.
J Neurosci ; 34(1): 295-304, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24381290

RESUMEN

Neuronal activity triggers endocytosis at synaptic terminals to retrieve efficiently the exocytosed vesicle membrane, ensuring the membrane homeostasis of active zones and the continuous supply of releasable vesicles. The kinetics of endocytosis depends on Ca(2+) and calmodulin which, as a versatile signal pathway, can activate a broad spectrum of downstream targets, including myosin light chain kinase (MLCK). MLCK is known to regulate vesicle trafficking and synaptic transmission, but whether this kinase regulates vesicle endocytosis at synapses remains elusive. We investigated this issue at the rat calyx of Held synapse, where previous studies using whole-cell membrane capacitance measurement have characterized two common forms of Ca(2+)/calmodulin-dependent endocytosis, i.e., slow clathrin-dependent endocytosis and rapid endocytosis. Acute inhibition of MLCK with pharmacological agents was found to slow down the kinetics of both slow and rapid forms of endocytosis at calyces. Similar impairment of endocytosis occurred when blocking myosin II, a motor protein that can be phosphorylated upon MLCK activation. The inhibition of endocytosis was not accompanied by a change in Ca(2+) channel current. Combined inhibition of MLCK and calmodulin did not induce synergistic inhibition of endocytosis. Together, our results suggest that activation of MLCK accelerates both slow and rapid forms of vesicle endocytosis at nerve terminals, likely by functioning downstream of Ca(2+)/calmodulin.


Asunto(s)
Tronco Encefálico/enzimología , Endocitosis/fisiología , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/metabolismo , Sinapsis/enzimología , Vesículas Sinápticas/enzimología , Animales , Tronco Encefálico/efectos de los fármacos , Endocitosis/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Femenino , Masculino , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley , Sinapsis/efectos de los fármacos , Vesículas Sinápticas/efectos de los fármacos
15.
Lab Invest ; 95(10): 1145-56, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26146960

RESUMEN

Hepatocellular carcinoma (HCC) carries a poor prognosis with no effective treatment available other than liver transplantation for selected patients. Vascular invasion of HCC is one of the most important negative predictor of survival. As the regulation of invasion of HCC cells is not well understood, our aim was to study the mechanisms by which galectin 3, a ß-galactosidase-binding lectin mediates HCC cell migration. HCC was induced by N-diethylnitrosamine in wild-type and galectin 3(-/-) mice, and tumor formation, histology, and tumor cell invasion were assessed. The galectin 3(-/-) mice developed significantly smaller tumor burden with a less invasive phenotype than the wild-type animals. Galectin 3 was upregulated in the wild-type HCC tumor tissue, but not in the surrounding parenchyma. Galectin 3 expression in HCC was induced by NF-κB transactivation as determined by chromatin immunoprecipitation assays. In vitro studies assessed the pro-migratory effects of galectin 3. The migration of hepatoma cells was significantly decreased after transfection by the galectin 3 siRNA and also after using the Rho kinase inhibitor Y-27632. The reorganization of the actin cytoskeleton, RhoA GTPase activity and the phosphorylation of MLC2 (myosin light chain 2) were decreased in the galectin 3 siRNA-transfected cells. In addition, in vitro and in vivo evidence showed that galectin 3 deficiency reduced hepatoma cell proliferation and increased their apoptosis rate. In conclusion, galectin 3 is an important lectin that is induced in HCC cells, and promotes hepatoma cell motility and invasion by an autocrine pathway. Targeting galectin 3 therefore could be an important novel treatment strategy to halt disease progression.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Galectina 3/metabolismo , Neoplasias Hepáticas/metabolismo , Hígado/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/patología , Animales , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Miosinas Cardíacas/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Galectina 3/antagonistas & inhibidores , Galectina 3/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Hígado/efectos de los fármacos , Hígado/patología , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/química , Invasividad Neoplásica , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/agonistas , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/genética
16.
Kidney Int ; 87(2): 370-81, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25140913

RESUMEN

Bolus administration of endothelin-1 elicits long-lasting renal afferent arteriolar vasoconstriction, in contrast to transient constriction induced by angiotensin II. Vasoconstriction is generally evoked by myosin regulatory light chain (LC20) phosphorylation at Ser19 by myosin light chain kinase (MLCK), which is enhanced by Rho-associated kinase (ROCK)-mediated inhibition of myosin light chain phosphatase (MLCP). LC20 can be diphosphorylated at Ser19 and Thr18, resulting in reduced rates of dephosphorylation and relaxation. Here we tested whether LC20 diphosphorylation contributes to sustained endothelin-1 but not transient angiotensin II-induced vasoconstriction. Endothelin-1 treatment of isolated arterioles elicited a concentration- and time-dependent increase in LC20 diphosphorylation at Thr18 and Ser19. Inhibition of MLCK or ROCK reduced endothelin-1-evoked LC20 mono- and diphosphorylation. Pretreatment with an ETB but not an ETA receptor antagonist abolished LC20 diphosphorylation, and an ETB receptor agonist induced LC20 diphosphorylation. In contrast, angiotensin II caused phosphorylation exclusively at Ser19. Thus, endothelin-1 and angiotensin II induce afferent arteriolar constriction via LC20 phosphorylation at Ser19 due to calcium activation of MLCK and ROCK-mediated inhibition of MLCP. Endothelin-1, but not angiotensin II, induces phosphorylation of LC20 at Thr18. This could contribute to the prolonged vasoconstrictor response to endothelin-1.


Asunto(s)
Angiotensina II/fisiología , Arteriolas/fisiología , Endotelina-1/fisiología , Cadenas Ligeras de Miosina/metabolismo , Vasoconstricción/fisiología , Angiotensina II/farmacología , Animales , Arteriolas/efectos de los fármacos , Bloqueadores de los Canales de Calcio/farmacología , Señalización del Calcio/efectos de los fármacos , Diltiazem/farmacología , Antagonistas de los Receptores de la Endotelina A/farmacología , Endotelina-1/farmacología , Modelos Cardiovasculares , Cadenas Ligeras de Miosina/química , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptor de Endotelina B/agonistas , Transducción de Señal/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
17.
Am J Physiol Gastrointest Liver Physiol ; 309(12): G988-97, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26514773

RESUMEN

Recent studies have implicated a pathogenic role for matrix metalloproteinases 9 (MMP-9) in inflammatory bowel disease. Although loss of epithelial barrier function has been shown to be a key pathogenic factor for the development of intestinal inflammation, the role of MMP-9 in intestinal barrier function remains unclear. The aim of this study was to investigate the role of MMP-9 in intestinal barrier function and intestinal inflammation. Wild-type (WT) and MMP-9(-/-) mice were subjected to experimental dextran sodium sulfate (DSS) colitis by administration of 3% DSS in drinking water for 7 days. The mouse colonic permeability was measured in vivo by recycling perfusion of the entire colon using fluorescently labeled dextran. The DSS-induced increase in the colonic permeability was accompanied by an increase in intestinal epithelial cell MMP-9 expression in WT mice. The DSS-induced increase in intestinal permeability and the severity of DSS colitis was found to be attenuated in MMP-9(-/-) mice. The colonic protein expression of myosin light chain kinase (MLCK) and phospho-MLC was found to be significantly increased after DSS administration in WT mice but not in MMP-9(-/-) mice. The DSS-induced increase in colonic permeability and colonic inflammation was attenuated in MLCK(-/-) mice and MLCK inhibitor ML-7-treated WT mice. The DSS-induced increase in colonic surface epithelial cell MLCK mRNA was abolished in MMP-9(-/-) mice. Lastly, increased MMP-9 protein expression was detected within the colonic surface epithelial cells in ulcerative colitis cases. These data suggest a role of MMP-9 in modulation of colonic epithelial permeability and inflammation via MLCK.


Asunto(s)
Colitis/enzimología , Colon/enzimología , Sulfato de Dextran , Mucosa Intestinal/enzimología , Metaloproteinasa 9 de la Matriz/metabolismo , Uniones Estrechas/enzimología , Animales , Colitis/inducido químicamente , Colitis/genética , Colitis/patología , Colitis/prevención & control , Colon/efectos de los fármacos , Colon/patología , Modelos Animales de Enfermedad , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Metaloproteinasa 9 de la Matriz/análisis , Metaloproteinasa 9 de la Matriz/deficiencia , Metaloproteinasa 9 de la Matriz/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/genética , Quinasa de Cadena Ligera de Miosina/metabolismo , Permeabilidad , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Índice de Severidad de la Enfermedad , Transducción de Señal , Proteínas de Uniones Estrechas/metabolismo , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/patología , Factores de Tiempo
18.
Biochem Biophys Res Commun ; 456(4): 884-90, 2015 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-25529450

RESUMEN

The 15-kDa selenoprotein (Sep15) has been implicated in etiology of some types of cancer. Herein, inducible RNAi cell lines were established and cell morphology and motility were analyzed. The majority of Sep15-deficient cells (>95%) formed membrane blebs in a dynamic manner. Blebbing cells transformed cell morphology from a normal flat spindle shape to a spherical morphology. In blebbing cells, actin fibers moved to the cell periphery, covering and obscuring visualization of α-tubulin. Bleb formation was suppressed by the inhibitors of Rho-associated protein kinase (ROCK), RhoA or myosin light chain (MLC), restoring blebbing cells to wild-type morphology. RhoA activation and phosphorylation of myosin phosphatase target subunit 1 was induced by Sep15 knockdown. Sep15-deficient cells were non-apoptotic, and displayed a distinct relative localization of F-actin and α-tubulin from typical apoptotic blebbing cells. Our data suggest that Sep15 in Chang liver cells regulates the pathway that antagonizes RhoA/ROCK/MLC-dependent non-apoptotic bleb formation.


Asunto(s)
Apoptosis , Estructuras de la Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Selenoproteínas/deficiencia , Transducción de Señal , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Amidas/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Estructuras de la Membrana Celular/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Piridinas/farmacología , Selenoproteínas/metabolismo , Transducción de Señal/efectos de los fármacos
19.
Basic Res Cardiol ; 110(4): 38, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25982880

RESUMEN

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.


Asunto(s)
Miosinas Cardíacas/fisiología , Glicoproteínas de Membrana/genética , Miocardio/metabolismo , Cadenas Ligeras de Miosina/fisiología , NADPH Oxidasas/genética , Animales , Miosinas Cardíacas/análisis , Núcleo Celular/química , Células Cultivadas , Masculino , Daño por Reperfusión Miocárdica/prevención & control , Cadenas Ligeras de Miosina/análisis , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , NADPH Oxidasa 2 , Estrés Oxidativo , Fosforilación , Ratas , Ratas Sprague-Dawley
20.
Mol Vis ; 21: 98-109, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25684975

RESUMEN

PURPOSE: Actin and myosin within the crystalline lens maintain the structural integrity of lens fiber cells and form a hexagonal lattice cradling the posterior surface of the lens. The actomyosin network was pharmacologically disrupted to examine the effects on lenticular biomechanics and optical quality. METHODS: One lens of 7-day-old White Leghorn chickens was treated with 10 µM of a disruptor and the other with 0.01% dimethyl sulfoxide (vehicle). Actin, myosin, and myosin light chain kinase (MLCK) disruptors were used. The stiffness and the optical quality of the control and treated lenses were measured. Western blotting and confocal imaging were used to confirm that treatment led to a disruption of the actomyosin network. The times for the lenses to recover stiffness to match the control values were also measured. RESULTS: Disruptor-treated lenses were significantly less stiff than their controls (p≤0.0274 for all disruptors). The disruptors led to changes in the relative protein amounts as well as the distributions of proteins within the lattice. However, the disruptors did not affect the clarity of the lenses (p≥0.4696 for all disruptors), nor did they affect spherical aberration (p = 0.02245). The effects of all three disruptors were reversible, with lenses recovering from treatment with actin, myosin, and MLCK disruptors after 4 h, 1 h, and 8 min, respectively. CONCLUSIONS: Cytoskeletal protein disruptors led to a decreased stiffness of the lens, and the effects were reversible. Optical quality was mostly unaffected, but the long-term consequences remain unclear. Our results raise the possibility that the mechanical properties of the avian lens may be actively regulated in vivo via adjustments to the actomyosin lattice.


Asunto(s)
Actomiosina/antagonistas & inhibidores , Azepinas/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Cristalino/efectos de los fármacos , Naftalenos/farmacología , Tiazolidinas/farmacología , Actinas/antagonistas & inhibidores , Actinas/metabolismo , Actomiosina/metabolismo , Animales , Animales Recién Nacidos , Proteínas Aviares/antagonistas & inhibidores , Proteínas Aviares/metabolismo , Pollos , Fuerza Compresiva/efectos de los fármacos , Dureza/efectos de los fármacos , Cristalino/metabolismo , Cristalino/ultraestructura , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/metabolismo , Miosinas/antagonistas & inhibidores , Miosinas/metabolismo , Técnicas de Cultivo de Tejidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA