Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Hum Mol Genet ; 27(9): 1608-1617, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29474540

RESUMEN

Centronuclear myopathies (CNM) are a subtype of congenital myopathies (CM) characterized by skeletal muscle weakness and an increase in the number of central myonuclei. We have previously identified three CNM probands, two with associated dilated cardiomyopathy, carrying striated preferentially expressed gene (SPEG) mutations. Currently, the role of SPEG in skeletal muscle function is unclear as constitutive SPEG-deficient mice developed severe dilated cardiomyopathy and died in utero. We have generated a conditional Speg-KO mouse model and excised Speg by crosses with striated muscle-specific cre-expressing mice (MCK-Cre). The resulting litters had a delay in Speg excision consistent with cre expression starting in early postnatal life and, therefore, an extended lifespan up to a few months. KO mice were significantly smaller and weaker than their littermate-matched controls. Histopathological skeletal muscle analysis revealed smaller myofibers, marked fiber-size variability, and poor integrity and low number of triads. Further, SPEG-deficient muscle fibers were weaker by physiological and in vitro studies and exhibited abnormal Ca2+ handling and excitation-contraction (E-C) coupling. Overall, SPEG deficiency in skeletal muscle is associated with fewer and abnormal triads, and defective calcium handling and excitation-contraction coupling, suggesting that therapies targeting calcium signaling may be beneficial in such patients.


Asunto(s)
Calcio/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Miopatías Estructurales Congénitas/metabolismo , Miopatías Estructurales Congénitas/patología , Quinasa de Cadena Ligera de Miosina/metabolismo , Animales , Señalización del Calcio/fisiología , Femenino , Ratones , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Quinasa de Cadena Ligera de Miosina/deficiencia , Quinasa de Cadena Ligera de Miosina/genética
2.
J Am Heart Assoc ; 7(3)2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29371201

RESUMEN

BACKGROUND: Obstructive sleep apnea is characterized by repetitive pharyngeal collapses during sleep, leading to intermittent hypoxia (IH), the main contributor of obstructive sleep apnea-related cardiovascular morbidity. In patients and rodents with obstructive sleep apnea exposed to IH, vascular inflammation and remodeling, endothelial dysfunction, and circulating inflammatory markers are linked with IH severity. The nonmuscle myosin light chain kinase (nmMLCK) isoform contributes to vascular inflammation and oxidative stress in different cardiovascular and inflammatory diseases. Thus, in the present study, we hypothesized that nmMLCK plays a key role in the IH-induced vascular dysfunctions and inflammatory remodeling. METHODS AND RESULTS: Twelve-week-old nmMLCK+/+ or nmMLCK-/- mice were exposed to 14-day IH or normoxia. IH was associated with functional alterations characterized by an elevation of arterial blood pressure and stiffness and perturbations of NO signaling. IH caused endothelial barrier dysfunction (ie, reduced transendothelial resistance in vitro) and induced vascular oxidative stress associated with an inflammatory remodeling, characterized by an increased intima-media thickness and an increased expression and activity of inflammatory markers, such as interferon-γ and nuclear factor-κB, in the vascular wall. Interestingly, nmMLCK deletion prevented all IH-induced functional and structural alterations, including the restoration of NO signaling, correction of endothelial barrier integrity, and reduction of both oxidative stress and associated inflammatory response. CONCLUSIONS: nmMLCK is a key mechanism in IH-induced vascular oxidative stress and inflammation and both functional and structural remodeling.


Asunto(s)
Arterias/enzimología , Hipoxia/complicaciones , Quinasa de Cadena Ligera de Miosina/metabolismo , Enfermedades Vasculares/etiología , Animales , Presión Arterial , Arterias/fisiopatología , Línea Celular , Modelos Animales de Enfermedad , Impedancia Eléctrica , Hipoxia/enzimología , Hipoxia/genética , Hipoxia/fisiopatología , Mediadores de Inflamación/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Quinasa de Cadena Ligera de Miosina/deficiencia , Quinasa de Cadena Ligera de Miosina/genética , Óxido Nítrico/metabolismo , Estrés Oxidativo , Transducción de Señal , Enfermedades Vasculares/enzimología , Enfermedades Vasculares/genética , Enfermedades Vasculares/fisiopatología , Remodelación Vascular , Rigidez Vascular
3.
Sci Rep ; 7(1): 13664, 2017 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-29057883

RESUMEN

Obstructive sleep apnea is characterized by intermittent hypoxia (IH) which alters endothelial function, induces inflammation and accelerates atherosclerosis-induced cardiovascular diseases. The non-muscular myosin light chain kinase (nmMLCK) isoform contributes to endothelial cell-cell junction opening. Deletion of nmMLCK protects mice from death in septic shock models and prevents atherosclerosis in high-fat diet-fed mice. The aim of the study was to analyze the implication of nmMLCK in IH-induced vascular inflammation. Human aortic endothelial cells were exposed to 6 hours of IH in absence or presence of nmMLCK inhibitors, ML-7 (5 µM) or PIK (150 µM). IH increased reactive oxygen species (ROS) and nitric oxide (NO) production, p65-NFκB activation and IL-6 secretion. While nmMLCK inhibition did not prevent IH-induced ROS production and p65-NFκB activation, it decreased NO production and partially prevented IL-6 secretion. IH-induced IL-6 secretion and vesicle-associated membrane protein-associated vesicles re-organization were inhibited in presence of the inhibitor of protein secretion, brefeldin A, or ML-7. IH increased monocytes transendothelial migration that was partially prevented by ML-7. Finally, IH reduced endothelium-dependent relaxation to acetylcholine of aortas from wild-type but not those taken from nmMLCK-deficient mice. These results suggest that nmMLCK participates to IH-induced endothelial dysfunction resulting from cytokines secretion and endothelial permeability.


Asunto(s)
Aorta/enzimología , Permeabilidad Capilar/fisiología , Hipoxia de la Célula/fisiología , Células Endoteliales/enzimología , Interleucina-6/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Animales , Aorta/efectos de los fármacos , Aorta/patología , Aterosclerosis/enzimología , Aterosclerosis/patología , Permeabilidad Capilar/efectos de los fármacos , Adhesión Celular/fisiología , Hipoxia de la Célula/efectos de los fármacos , Movimiento Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Humanos , Ratones Noqueados , Quinasa de Cadena Ligera de Miosina/deficiencia , Quinasa de Cadena Ligera de Miosina/genética , Especies Reactivas de Oxígeno/metabolismo , Vesículas Secretoras/efectos de los fármacos , Vesículas Secretoras/enzimología , Vesículas Secretoras/patología , Apnea Obstructiva del Sueño/enzimología , Factor de Transcripción ReIA/metabolismo
4.
Nat Commun ; 7: 11358, 2016 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-27101932

RESUMEN

Smooth muscle sphincters exhibit basal tone and control passage of contents through organs such as the gastrointestinal tract; loss of this tone leads to disorders such as faecal incontinence. However, the molecular mechanisms underlying this tone remain unknown. Here, we show that deletion of myosin light-chain kinases (MLCK) in the smooth muscle cells from internal anal sphincter (IAS-SMCs) abolishes basal tone, impairing defecation. Pharmacological regulation of ryanodine receptors (RyRs), L-type voltage-dependent Ca(2+) channels (VDCCs) or TMEM16A Ca(2+)-activated Cl(-) channels significantly changes global cytosolic Ca(2+) concentration ([Ca(2+)]i) and the tone. TMEM16A deletion in IAS-SMCs abolishes the effects of modulators for TMEM16A or VDCCs on a RyR-mediated rise in global [Ca(2+)]i and impairs the tone and defecation. Hence, MLCK activation in IAS-SMCs caused by a global rise in [Ca(2+)]i via a RyR-TMEM16A-VDCC signalling module sets the basal tone. Targeting this module may lead to new treatments for diseases like faecal incontinence.


Asunto(s)
Canal Anal/metabolismo , Canales de Calcio Tipo L/metabolismo , Canales de Cloruro/metabolismo , Incontinencia Fecal/metabolismo , Hipotonía Muscular/metabolismo , Quinasa de Cadena Ligera de Miosina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Canal Anal/efectos de los fármacos , Canal Anal/fisiopatología , Animales , Anoctamina-1 , Betanecol/farmacología , Calcio/metabolismo , Canales de Calcio Tipo L/genética , Señalización del Calcio , Canales de Cloruro/genética , Defecación/efectos de los fármacos , Incontinencia Fecal/genética , Incontinencia Fecal/fisiopatología , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Contracción Muscular/efectos de los fármacos , Hipotonía Muscular/genética , Hipotonía Muscular/fisiopatología , Músculo Liso/efectos de los fármacos , Músculo Liso/metabolismo , Músculo Liso/fisiopatología , Quinasa de Cadena Ligera de Miosina/deficiencia , Nifedipino/farmacología , Ácido Niflúmico/farmacología , Técnicas de Placa-Clamp , Canal Liberador de Calcio Receptor de Rianodina/genética
5.
Cardiovasc Res ; 111(1): 34-43, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27025239

RESUMEN

AIMS: Under pressure overload, initial adaptive hypertrophy of the heart is followed by cardiomyocyte elongation, reduced contractile force, and failure. The mechanisms governing the transition to failure are not fully understood. Pressure overload reduced cardiac myosin light chain kinase (cMLCK) by ∼80% within 1 week and persists. Knockdown of cMLCK in cardiomyocytes resulted in reduced cardiac contractility and sarcomere disorganization. Thus, we hypothesized that acute reduction of cMLCK may be causative for reduced contractility and cardiomyocyte remodelling during the transition from compensated to decompensated cardiac hypertrophy. METHODS AND RESULTS: To mimic acute cMLCK reduction in adult hearts, the floxed-Mylk3 gene that encodes cMLCK was inducibly ablated in Mylk3(flox/flox)/merCremer mice (Mylk3-KO), and compared with two control mice (Mylk3(flox/flox) and Mylk3(+/+)/merCremer) following tamoxifen injection (50 mg/kg/day, 2 consecutive days). In Mylk3-KO mice, reduction of cMLCK protein was evident by 4 days, with a decline to below the level of detection by 6 days. By 7 days, these mice exhibited heart failure, with reduction of fractional shortening compared with those in two control groups (19.8 vs. 28.0% and 27.7%). Severely convoluted cardiomyocytes with sarcomeric disorganization, wavy fibres, and cell death were demonstrated in Mylk3-KO mice. The cardiomyocytes were also unable to thicken adaptively to pressure overload. CONCLUSION: Our results, using a new mouse model mimicking an acute reduction of cMLCK, suggest that cMLCK plays a pivotal role in the transition from compensated to decompensated hypertrophy via sarcomeric disorganization.


Asunto(s)
Cardiomegalia/enzimología , Insuficiencia Cardíaca/enzimología , Miocitos Cardíacos/enzimología , Quinasa de Cadena Ligera de Miosina/deficiencia , Función Ventricular Izquierda , Remodelación Ventricular , Enfermedad Aguda , Adaptación Fisiológica , Animales , Atrofia , Señalización del Calcio , Miosinas Cardíacas/metabolismo , Cardiomegalia/genética , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Predisposición Genética a la Enfermedad , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Contracción Miocárdica , Miocitos Cardíacos/patología , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/genética , Fenotipo , Fosforilación , Sarcómeros/enzimología , Sarcómeros/patología , Factores de Tiempo
6.
Muscle Nerve ; 54(2): 308-16, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26802366

RESUMEN

INTRODUCTION: Posttetanic potentiation (PTP) and the catchlike property (CLP) enhance contractile function in skeletal muscle. We investigated the CLP during dynamic performance in mouse hindlimb muscles with (wild-type) and without (skMLCK(-/-) ) the primary mechanism for PTP (myosin phosphorylation) (in vitro, 25°C). METHODS: Extensor digitorum longus muscles of both genotypes were stimulated with constant frequency and catchlike trains (CFT and CLT), before and after a potentiating stimulus (PS). RESULTS: Before the PS, the CLT increased concentric force/work relative to the CFT, but this effect was greater for skMLCK(-/-) than wild-type muscles. After the PS, the catchlike effect was reduced in wild-type muscles but unchanged in skMLCK(-/-) muscles that did not display PTP. CONCLUSIONS: These data suggest that PTP interferes with the CLP during concentric force development at moderate speeds of shortening. We conclude that the physiological utility of each mechanism and their interactions provide important modulations to fast skeletal muscle function. Muscle Nerve 54: 308-316, 2016.


Asunto(s)
Potenciales de Acción/fisiología , Estimulación Eléctrica , Contracción Muscular/fisiología , Músculo Esquelético/fisiología , Potenciales de Acción/genética , Animales , Fenómenos Biomecánicos , Electromiografía , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Contracción Muscular/genética , Músculo Esquelético/inervación , Quinasa de Cadena Ligera de Miosina/deficiencia , Quinasa de Cadena Ligera de Miosina/genética , Fosforilación
7.
Am J Physiol Heart Circ Physiol ; 308(9): H1039-50, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25724497

RESUMEN

Microcirculatory dysfunction may cause tissue malperfusion and progression to organ failure in the later stages of sepsis, but the role of smooth muscle contractile dysfunction is uncertain. Mice were given intraperitoneal LPS, and mesenteric arteries were harvested at 6-h intervals for analyses of gene expression and contractile function by wire myography. Contractile (myosin and actin) and regulatory [myosin light chain kinase and phosphatase subunits (Mypt1, CPI-17)] mRNAs and proteins were decreased in mesenteric arteries at 24 h concordant with reduced force generation to depolarization, Ca(2+), and phenylephrine. Vasodilator sensitivity to DEA/nitric oxide (NO) and cGMP under Ca(2+) clamp were increased at 24 h after LPS concordant with a switch to Mypt1 exon 24- splice variant coding for a leucine zipper (LZ) motif required for PKG-1α activation of myosin phosphatase. This was reproduced by smooth muscle-specific deletion of Mypt1 exon 24, causing a shift to the Mypt1 LZ+ isoform. These mice had significantly lower resting blood pressure than control mice but similar hypotensive responses to LPS. The vasodilator sensitivity of wild-type mice to DEA/NO, but not cGMP, was increased at 6 h after LPS. This was abrogated in mice with a redox dead version of PKG-1α (Cys42Ser). Enhanced vasorelaxation in early endotoxemia is mediated by redox signaling through PKG-1α but in later endotoxemia by myosin phosphatase isoform shifts enhancing sensitivity to NO/cGMP as well as smooth muscle atrophy. Muscle atrophy and modulation may be a novel target to suppress microcirculatory dysfunction; however, inactivation of inducible NO synthase, treatment with the IL-1 antagonist IL-1ra, or early activation of α-adrenergic signaling did not suppressed this response.


Asunto(s)
Lipopolisacáridos , Proteínas Musculares/metabolismo , Músculo Liso Vascular/enzimología , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Óxido Nítrico/metabolismo , Fosfoproteínas/metabolismo , Sepsis/enzimología , Transducción de Señal , Vasodilatación , Animales , GMP Cíclico/metabolismo , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/deficiencia , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/genética , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica , Genotipo , Péptidos y Proteínas de Señalización Intracelular , Isoenzimas , Masculino , Arterias Mesentéricas/enzimología , Arterias Mesentéricas/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Microcirculación , Proteínas Musculares/genética , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiopatología , Atrofia Muscular/inducido químicamente , Atrofia Muscular/enzimología , Atrofia Muscular/fisiopatología , Quinasa de Cadena Ligera de Miosina/deficiencia , Quinasa de Cadena Ligera de Miosina/genética , Fosfatasa de Miosina de Cadena Ligera/genética , Óxido Nítrico Sintasa de Tipo II/deficiencia , Óxido Nítrico Sintasa de Tipo II/genética , Oxidación-Reducción , Fenotipo , Fosfoproteínas/genética , ARN Mensajero/metabolismo , Sepsis/inducido químicamente , Sepsis/genética , Sepsis/fisiopatología , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Vasoconstrictores/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
8.
Can J Physiol Pharmacol ; 93(1): 23-32, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25412230

RESUMEN

The isometric potentiation associated with myosin phosphorylation is force dependent. The purpose of this study was to assess the influence of a pre-existing period of isometric force on the concentric force potentiation displayed by mouse muscles with and without the ability to phosphorylate myosin. We tested isometric (ISO) and concentric (CON) potentiation, as well as concentric potentiation after isometric force (ISO-CON), in muscles from wild-type (WT) and skeletal myosin light chain kinase-deficient (skMLCK(-/-)) mice. A conditioning stimulus increased (i.e., potentiated) mean concentric force in the ISO-CON and CON conditions to 1.31 ± 0.02 and 1.35 ± 0.02 (WT) and to 1.19 ± 0.02 and 1.21 ± 0.01 (skMLCK(-/-)) of prestimulus levels, respectively (data n = 6-8, p < 0.05). No potentiation of mean isometric force was observed in either genotype. The potentiation of mean concentric force was inversely related to relative tetanic force level (P/Po) in both genotypes. Moreover, concentric potentiation varied greatly within each contraction type and was negatively correlated with unpotentiated force in both genotypes. Thus, although no effect of pre-existing force was observed, strong and inverse relationships between concentric force potentiation and unpotentiated concentric force may suggest an influence of attached and force-generating crossbridges on potentiation magnitude in both WT and skMLCK(-/-) muscles.


Asunto(s)
Contracción Isométrica/fisiología , Músculo Esquelético/enzimología , Quinasa de Cadena Ligera de Miosina/deficiencia , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Quinasa de Cadena Ligera de Miosina/metabolismo
9.
J Biol Chem ; 289(32): 22512-23, 2014 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-24951589

RESUMEN

Myosin light chain phosphatase with its regulatory subunit, myosin phosphatase target subunit 1 (MYPT1) modulates Ca(2+)-dependent phosphorylation of myosin light chain by myosin light chain kinase, which is essential for smooth muscle contraction. The role of MYPT1 in vascular smooth muscle was investigated in adult MYPT1 smooth muscle specific knock-out mice. MYPT1 deletion enhanced phosphorylation of myosin regulatory light chain and contractile force in isolated mesenteric arteries treated with KCl and various vascular agonists. The contractile responses of arteries from knock-out mice to norepinephrine were inhibited by Rho-associated kinase (ROCK) and protein kinase C inhibitors and were associated with inhibition of phosphorylation of the myosin light chain phosphatase inhibitor CPI-17. Additionally, stimulation of the NO/cGMP/protein kinase G (PKG) signaling pathway still resulted in relaxation of MYPT1-deficient mesenteric arteries, indicating phosphorylation of MYPT1 by PKG is not a major contributor to the relaxation response. Thus, MYPT1 enhances myosin light chain phosphatase activity sufficient for blood pressure maintenance. Rho-associated kinase phosphorylation of CPI-17 plays a significant role in enhancing vascular contractile responses, whereas phosphorylation of MYPT1 in the NO/cGMP/PKG signaling module is not necessary for relaxation.


Asunto(s)
Músculo Liso Vascular/fisiología , Quinasa de Cadena Ligera de Miosina/fisiología , Animales , Presión Sanguínea/fisiología , Femenino , Hipertensión/etiología , Hipertensión/fisiopatología , Péptidos y Proteínas de Señalización Intracelular , Masculino , Arterias Mesentéricas/fisiología , Ratones , Ratones Noqueados , Proteínas Musculares/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/deficiencia , Quinasa de Cadena Ligera de Miosina/genética , Fosfatasa de Miosina de Cadena Ligera , Óxido Nítrico/metabolismo , Fosfoproteínas/metabolismo , Fosforilación , Transducción de Señal , Vasoconstricción/fisiología , Vasodilatación/fisiología
10.
Int J Mol Med ; 33(4): 905-12, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24481113

RESUMEN

Myosin light chain kinase (MLCK) phosphorylates myosin regulatory light chains to facilitate its interaction with actin filaments and produce contractile activity. The outer hair cells (OHCs) in the ear contain large amounts of actin and a variety myosins. The stereociliary and somatic motility of OHCs are closely related to hearing. It appears likely that MLCK may play an important role in acoustic trans-duction. In this study, we analyzed, both in vivo and in vitro, the OHCs of mice bearing a specific deletion of the MLCK gene and the OHCs of control mice. The phenotype was assessed by auditory function [acoustic brainstem responses (ABRs) and distortion product otoacoustic emissions (DPOAEs)], inner ear morphology and histology. MLCK-deficient mice aged 6-7 months showed impaired hearing, a 5- to 10-dB sound pressure level (SPL) increase in the ABR thresholds, when responding to clicks and tones of different frequencies (8 and 16 kHz) (P<0.05). The DPOAE amplitudes of 3-month-old MLCK-deficient mice decreased significantly (>10 dB SPL) at low frequencies (4, 5 and 6 kHz). The OHCs in the MLCK-deficient mice increased with abnormal stereocilia. The staining of F-actin and the phosphorylation of the regulatory light chain in MLCK-deficient OHCs was weak. Our results indicate that MLCK may regulate the structure and the motility of stereocilia through F-actin polymerization.


Asunto(s)
Actinas/metabolismo , Citoesqueleto/metabolismo , Células Ciliadas Auditivas Externas/enzimología , Células Ciliadas Auditivas Externas/fisiología , Audición/fisiología , Quinasa de Cadena Ligera de Miosina/metabolismo , Animales , Umbral Auditivo/fisiología , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Técnica del Anticuerpo Fluorescente , Células Ciliadas Auditivas Externas/citología , Células Ciliadas Auditivas Externas/ultraestructura , Ratones , Ratones Noqueados , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/deficiencia , Emisiones Otoacústicas Espontáneas/fisiología , Fosforilación
11.
PLoS One ; 8(6): e66720, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23776695

RESUMEN

BACKGROUND: Activation of ErbB2/4 receptor tyrosine kinases in cardiomyocytes by neuregulin treatment is associated with improvement in cardiac function, supporting its use in human patients with heart failure despite the lack of a specific mechanism. Neuregulin infusion in rodents increases cardiac myosin light chain kinase (cMLCK) expression and cardiac myosin regulatory light chain (RLC) phosphorylation which may improve actin-myosin interactions for contraction. We generated a cMLCK knockout mouse to test the hypothesis that cMLCK is necessary for neuregulin-induced improvement in cardiac function by increasing RLC phosphorylation. PRINCIPAL FINDINGS: The cMLCK knockout mice have attenuated RLC phosphorylation and decreased cardiac performance measured as fractional shortening. Neuregulin infusion for seven days in wildtype mice increased cardiac cMLCK protein expression and RLC phosphorylation while increasing Akt phosphorylation and decreasing phospholamban phosphorylation. There was no change in fractional shortening. In contrast, neuregulin infusion in cMLCK knockout animals increased cardiac performance in the absence of cMLCK without increasing RLC phosphorylation. In addition, CaMKII signaling appeared to be enhanced in neuregulin-treated knockout mice. CONCLUSIONS: Thus, Neuregulin may improve cardiac performance in the failing heart without increasing cMLCK and RLC phosphorylation by activating other signaling pathways.


Asunto(s)
Corazón/efectos de los fármacos , Quinasa de Cadena Ligera de Miosina/deficiencia , Quinasa de Cadena Ligera de Miosina/metabolismo , Neurregulinas/farmacología , Animales , Masculino , Ratones , Ratones Noqueados , Miocardio/metabolismo , Miocardio/patología , Quinasa de Cadena Ligera de Miosina/genética , Fosforilación/efectos de los fármacos , ARN Mensajero/genética , Transducción de Señal/efectos de los fármacos
12.
Gastroenterology ; 144(7): 1456-65, 1465.e1-5, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23499953

RESUMEN

BACKGROUND & AIMS: The regulatory subunit of myosin light chain phosphatase, MYPT1, has been proposed to control smooth muscle contractility by regulating phosphorylation of the Ca(2+)-dependent myosin regulatory light chain. We generated mice with a smooth muscle-specific deletion of MYPT1 to investigate its physiologic role in intestinal smooth muscle contraction. METHODS: We used the Cre-loxP system to establish Mypt1-floxed mice, with the promoter region and exon 1 of Mypt1 flanked by 2 loxP sites. These mice were crossed with SMA-Cre transgenic mice to generate mice with smooth muscle-specific deletion of MYPT1 (Mypt1(SMKO) mice). The phenotype was assessed by histologic, biochemical, molecular, and physiologic analyses. RESULTS: Young adult Mypt1(SMKO) mice had normal intestinal motility in vivo, with no histologic abnormalities. On stimulation with KCl or acetylcholine, intestinal smooth muscles isolated from Mypt1(SMKO) mice produced robust and increased sustained force due to increased phosphorylation of the myosin regulatory light chain compared with muscle from control mice. Additional analyses of contractile properties showed reduced rates of force development and relaxation, and decreased shortening velocity, compared with muscle from control mice. Permeable smooth muscle fibers from Mypt1(SMKO) mice had increased sensitivity and contraction in response to Ca(2+). CONCLUSIONS: MYPT1 is not essential for smooth muscle function in mice but regulates the Ca(2+) sensitivity of force development and contributes to intestinal phasic contractile phenotype. Altered contractile responses in isolated tissues could be compensated by adaptive physiologic responses in vivo, where gut motility is affected by lower intensities of smooth muscle stimulation for myosin phosphorylation and force development.


Asunto(s)
Señalización del Calcio/fisiología , Motilidad Gastrointestinal/fisiología , Intestinos/fisiología , Contracción Muscular/fisiología , Músculo Liso/fisiología , Quinasa de Cadena Ligera de Miosina/fisiología , Animales , Calcio/metabolismo , Señalización del Calcio/genética , Femenino , Motilidad Gastrointestinal/genética , Masculino , Ratones , Ratones Noqueados , Contracción Muscular/genética , Quinasa de Cadena Ligera de Miosina/deficiencia , Quinasa de Cadena Ligera de Miosina/genética , Fosfatasa de Miosina de Cadena Ligera
13.
PLoS One ; 7(4): e34894, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22485190

RESUMEN

The structural homeostasis of the cochlear hair cell membrane is critical for all aspects of sensory transduction, but the regulation of its maintenance is not well understood. In this report, we analyzed the cochlear hair cells of mice with specific deletion of myosin light chain kinase (MLCK) in inner hair cells. MLCK-deficient mice showed impaired hearing, with a 5- to 14-dB rise in the auditory brainstem response (ABR) thresholds to clicks and tones of different frequencies and a significant decrease in the amplitude of the ABR waves. The mutant inner hair cells produced several ball-like structures around the hair bundles in vivo, indicating impaired membrane stability. Inner hair cells isolated from the knockout mice consistently displayed less resistance to hypoosmotic solution and less membrane F-actin. Myosin light-chain phosphorylation was also reduced in the mutated inner hair cells. Our results suggest that MLCK is necessary for maintaining the membrane stability of inner hair cells.


Asunto(s)
Membrana Celular/enzimología , Células Ciliadas Auditivas Internas/enzimología , Homeostasis , Quinasa de Cadena Ligera de Miosina/fisiología , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animales , Membrana Celular/metabolismo , Epitelio/enzimología , Epitelio/metabolismo , Potenciales Evocados Auditivos del Tronco Encefálico , Expresión Génica , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Internas/ultraestructura , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Cadenas Ligeras de Miosina/metabolismo , Miosina VIIa , Quinasa de Cadena Ligera de Miosina/deficiencia , Quinasa de Cadena Ligera de Miosina/genética , Miosinas/metabolismo , Órgano Espiral/citología , Presión Osmótica , Fosforilación , Procesamiento Proteico-Postraduccional , Eliminación de Secuencia , ATPasa Intercambiadora de Sodio-Potasio/genética , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
14.
Circulation ; 124(1): 48-57, 2011 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-21670231

RESUMEN

BACKGROUND: Endothelial dysfunction and monocyte migration are key events in the pathogenesis of atherosclerosis. Nonmuscle myosin light-chain kinase (nmMLCK), the predominant MLCK isoform in endothelial cells, has been shown to contribute to vascular inflammation by altering endothelial barrier function. However, its impact on atherogenesis remains unknown. METHODS AND RESULTS: We investigated the role of nmMLCK in the development of atherosclerotic lesions in apolipoprotein E-deficient (apoE(-/-)) mice fed an atherogenic diet for 12 weeks. Histopathological examination demonstrated that nmMLCK deficiency (apoE(-/-)nmmlck(-/-)) reduced the size of aortic lesions by 53%, lipid contents by 44%, and macrophage deposition by 40%. Western blotting and reverse-transcription polymerase chain reaction revealed the expression of nmMLCK in aortic endothelial cells and peripheral blood monocytes. Measurements of transendothelial electric resistance indicated that nmMLCK deficiency attenuated endothelial barrier dysfunction caused by thrombin, oxidized low-density lipoprotein, and tumor necrosis factor α. In monocytes, nmMLCK deficiency reduced their migration in response to the chemokine monocyte chemoattractant protein-1. Further mechanistic studies showed that nmMLCK acted through both myosin light chain phosphorylation-coupled and -uncoupled pathways; the latter involved Rous sacracoma virus homolog genes-encoded tyrosine kinases (Src) signaling. Moreover, depletion of Src via gene silencing, site-specific mutagenesis, or pharmacological inhibition of Src greatly attenuated nmMLCK-dependent endothelial barrier dysfunction and monocyte migration. CONCLUSIONS: Nonmuscle myosin light-chain kinase contributes to atherosclerosis by regulating endothelial barrier function and monocyte migration via mechanisms involving not only kinase-mediated MLC phosphorylation but also Src activation.


Asunto(s)
Apolipoproteínas E/deficiencia , Aterosclerosis/prevención & control , Movimiento Celular/fisiología , Endotelio Vascular/fisiopatología , Monocitos/fisiología , Quinasa de Cadena Ligera de Miosina/deficiencia , Animales , Aorta/patología , Apolipoproteínas E/genética , Apolipoproteínas E/fisiología , Aterosclerosis/patología , Aterosclerosis/fisiopatología , Células Cultivadas , Colágeno/metabolismo , Modelos Animales de Enfermedad , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Metabolismo de los Lípidos/fisiología , Macrófagos/patología , Masculino , Ratones , Ratones Noqueados , Monocitos/patología , Quinasa de Cadena Ligera de Miosina/genética , Quinasa de Cadena Ligera de Miosina/fisiología , Fosforilación , Transducción de Señal/fisiología , Túnica Íntima/metabolismo , Familia-src Quinasas/fisiología
15.
Am J Physiol Heart Circ Physiol ; 301(2): H584-91, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21572007

RESUMEN

Vascular tone, an important determinant of systemic vascular resistance and thus blood pressure, is affected by vascular smooth muscle (VSM) contraction. Key signaling pathways for VSM contraction converge on phosphorylation of the regulatory light chain (RLC) of smooth muscle myosin. This phosphorylation is mediated by Ca(2+)/calmodulin-dependent myosin light chain kinase (MLCK) but Ca(2+)-independent kinases may also contribute, particularly in sustained contractions. Signaling through MLCK has been indirectly implicated in maintenance of basal blood pressure, whereas signaling through RhoA has been implicated in salt-induced hypertension. In this report, we analyzed mice with smooth muscle-specific knockout of MLCK. Mesenteric artery segments isolated from smooth muscle-specific MLCK knockout mice (MLCK(SMKO)) had a significantly reduced contractile response to KCl and vasoconstrictors. The kinase knockout also markedly reduced RLC phosphorylation and developed force. We suggest that MLCK and its phosphorylation of RLC are required for tonic VSM contraction. MLCK(SMKO) mice exhibit significantly lower basal blood pressure and weaker responses to vasopressors. The elevated blood pressure in salt-induced hypertension is reduced below normotensive levels after MLCK attenuation. These results suggest that MLCK is necessary for both physiological and pathological blood pressure. MLCK(SMKO) mice may be a useful model of vascular failure and hypotension.


Asunto(s)
Presión Sanguínea , Hipertensión/enzimología , Músculo Liso Vascular/enzimología , Quinasa de Cadena Ligera de Miosina/metabolismo , Cloruro de Sodio Dietético , Vasoconstricción , Animales , Presión Sanguínea/efectos de los fármacos , Desoxicorticosterona , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Genotipo , Hipertensión/etiología , Hipertensión/fisiopatología , Arterias Mesentéricas/enzimología , Arterias Mesentéricas/fisiopatología , Ratones , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiopatología , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/deficiencia , Quinasa de Cadena Ligera de Miosina/genética , Nefrectomía , Fenotipo , Fosforilación , Cloruro de Potasio/farmacología , Factores de Tiempo , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología
16.
J Muscle Res Cell Motil ; 31(5-6): 337-48, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21298329

RESUMEN

Contraction-induced activation of a skeletal muscle specific Ca(2+) and calmodulin dependent myosin light chain kinase (skMLCK) catalyzes phosphorylation of the myosin regulatory light chain (RLC), a reaction that potentiates twitch force. The purpose of this study was to test the effect of skMLCK gene ablation on the fatigability of mouse extensor digitorum longus (EDL) muscle (in vitro at 25°C). Muscles were isolated from wildtype (WT, n = 10-12) and skeletal MLCK knockout (skMLCK KO, n = 10-12) mice and fatigued using a protocol consisting of 5 min of repeated tetanic stimulation (150 Hz for 1000 ms every 5 s). Both twitch (P(t)) and tetanic (P(o)) force as well as unloaded shortening velocity (V(o)) were assessed before, during and after fatiguing stimulation. Fatiguing stimulation increased RLC phosphorylation in WT but not skMLCK KO muscles (16 ± 0.01-0.63 ± 0.02 and 0.07 ± 0.02-0.08 ± 0.02 mol phos mol RLC, respectively). Although P(t) was potentiated above baseline in both WT and KO muscles, this increase was greater in WT than in KO muscles (to 1.37 ± 0.05 vs. 1.14 ± 0.02 of unpotentiated values, respectively). The difference in P(t) persisted until P(o) had been diminished to ~60% of baseline and thereafter P(t) declined to similar levels in both WT and KO muscles (to ~35% of initial). Overall, the time-course and decline in P(o) for WT and KO was similar (reduced to 0.20 ± 0.01 and 0.20 ± 0.01 of baseline, respectively) (P < 0.05). Initial values for V(o) were similar between WT and KO muscles and, moreover, the fatigue related decline in Vo was similar for both muscle genotypes (P < 0.05). Thus, our results demonstrate that skMLCK--catalyzed RLC phosphorylation augments isometric twitch force during moderate, but not severe, levels of fatigue.


Asunto(s)
Fatiga Muscular/genética , Quinasa de Cadena Ligera de Miosina/deficiencia , Quinasa de Cadena Ligera de Miosina/genética , Animales , Biocatálisis , Eliminación de Gen , Contracción Isométrica/genética , Contracción Isométrica/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fatiga Muscular/fisiología , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Fosforilación
17.
Am J Respir Cell Mol Biol ; 44(1): 40-52, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20139351

RESUMEN

Acute lung injury (ALI) and mechanical ventilator-induced lung injury (VILI), major causes of acute respiratory failure with elevated morbidity and mortality, are characterized by significant pulmonary inflammation and alveolar/vascular barrier dysfunction. Previous studies highlighted the role of the non-muscle myosin light chain kinase isoform (nmMLCK) as an essential element of the inflammatory response, with variants in the MYLK gene that contribute to ALI susceptibility. To define nmMLCK involvement further in acute inflammatory syndromes, we used two murine models of inflammatory lung injury, induced by either an intratracheal administration of lipopolysaccharide (LPS model) or mechanical ventilation with increased tidal volumes (the VILI model). Intravenous delivery of the membrane-permeant MLC kinase peptide inhibitor, PIK, produced a dose-dependent attenuation of both LPS-induced lung inflammation and VILI (~50% reductions in alveolar/vascular permeability and leukocyte influx). Intravenous injections of nmMLCK silencing RNA, either directly or as cargo within angiotensin-converting enzyme (ACE) antibody-conjugated liposomes (to target the pulmonary vasculature selectively), decreased nmMLCK lung expression (∼70% reduction) and significantly attenuated LPS-induced and VILI-induced lung inflammation (∼40% reduction in bronchoalveolar lavage protein). Compared with wild-type mice, nmMLCK knockout mice were significantly protected from VILI, with significant reductions in VILI-induced gene expression in biological pathways such as nrf2-mediated oxidative stress, coagulation, p53-signaling, leukocyte extravasation, and IL-6-signaling. These studies validate nmMLCK as an attractive target for ameliorating the adverse effects of dysregulated lung inflammation.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Terapia Genética/métodos , Pulmón/efectos de los fármacos , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , Lesión Pulmonar Inducida por Ventilación Mecánica/prevención & control , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/enzimología , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/inmunología , Animales , Anticuerpos , Líquido del Lavado Bronquioalveolar/inmunología , Permeabilidad Capilar/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Mediadores de Inflamación/metabolismo , Inyecciones Intravenosas , Lipopolisacáridos , Liposomas , Pulmón/irrigación sanguínea , Pulmón/enzimología , Pulmón/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/deficiencia , Quinasa de Cadena Ligera de Miosina/genética , Quinasa de Cadena Ligera de Miosina/metabolismo , Peptidil-Dipeptidasa A/inmunología , Fosforilación , Inhibidores de Proteínas Quinasas/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Transducción de Señal/genética , Factores de Tiempo , Lesión Pulmonar Inducida por Ventilación Mecánica/enzimología , Lesión Pulmonar Inducida por Ventilación Mecánica/genética , Lesión Pulmonar Inducida por Ventilación Mecánica/inmunología
18.
Proc Natl Acad Sci U S A ; 107(18): 8237-41, 2010 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-20404178

RESUMEN

The perijunctional actomyosin ring contributes to myosin light chain kinase (MLCK)-dependent tight junction regulation. However, the specific protein interactions involved in this process are unknown. To test the hypothesis that molecular remodeling contributes to barrier regulation, tight junction protein dynamic behavior was assessed by fluorescence recovery after photobleaching (FRAP). MLCK inhibition increased barrier function and stabilized ZO-1 at the tight junction but did not affect claudin-1, occludin, or actin exchange in vitro. Pharmacologic MLCK inhibition also blocked in vivo ZO-1 exchange in wild-type, but not long MLCK(-/-), mice. Conversely, ZO-1 exchange was accelerated in transgenic mice expressing constitutively active MLCK. In vitro, ZO-1 lacking the actin binding region (ABR) was not stabilized by MLCK inhibition, either in the presence or absence of endogenous ZO-1. Moreover, the free ABR interfered with full-length ZO-1 exchange and reduced basal barrier function. The free ABR also prevented increases in barrier function following MLCK inhibition in a manner that required endogenous ZO-1 expression. In silico modeling of the FRAP data suggests that tight junction-associated ZO-1 exists in three pools, two of which exchange with cytosolic ZO-1. Transport of the ABR-anchored exchangeable pool is regulated by MLCK. These data demonstrate a critical role for the ZO-1 ABR in barrier function and suggest that MLCK-dependent ZO-1 exchange is essential to this mechanism of barrier regulation.


Asunto(s)
Actinas/metabolismo , Proteínas de la Membrana/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Fosfoproteínas/metabolismo , Uniones Estrechas/metabolismo , Animales , Sitios de Unión , Células CACO-2 , Humanos , Ratones , Ratones Noqueados , Quinasa de Cadena Ligera de Miosina/deficiencia , Unión Proteica , Transporte de Proteínas , Proteína de la Zonula Occludens-1
19.
Circulation ; 119(2): 261-8, 2009 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-19118250

RESUMEN

BACKGROUND: The striated preferentially expressed gene (Speg) generates 4 different isoforms through alternative promoter use and tissue-specific splicing. Depending on the cell type, Speg isoforms may serve as markers of striated or smooth muscle differentiation. METHODS AND RESULTS: To elucidate function of Speg gene isoforms, we disrupted the Speg gene locus in mice by replacing common exons 8, 9, and 10 with a lacZ gene. beta-Galactosidase activity was detected in cardiomyocytes of the developing heart starting at day 11.5 days post coitum (dpc). beta-Galactosidase activity in other cell types, including vascular smooth muscle cells, did not begin until 18.5 dpc. In the developing heart, protein expression of only Spegalpha and Spegbeta isoforms was present in cardiomyocytes. Homozygous Speg mutant hearts began to enlarge by 16.5 dpc, and by 18.5 dpc, they demonstrated dilation of right and left atria and ventricles. These cardiac abnormalities in the absence of Speg were associated with a cellular hypertrophic response, myofibril degeneration, and a marked decrease in cardiac function. Moreover, Speg mutant mice exhibited significant neonatal mortality, with increased death occurring by 2 days after birth. CONCLUSIONS: These findings demonstrate that mutation of the Speg locus leads to cardiac dysfunction and a phenotype consistent with a dilated cardiomyopathy.


Asunto(s)
Cardiomiopatía Dilatada/enzimología , Cardiomiopatía Dilatada/genética , Proteínas Musculares/biosíntesis , Proteínas Musculares/genética , Quinasa de Cadena Ligera de Miosina/biosíntesis , Quinasa de Cadena Ligera de Miosina/genética , Animales , Animales Recién Nacidos , Cardiomiopatía Dilatada/etiología , Marcación de Gen , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Musculares/deficiencia , Mutagénesis Sitio-Dirigida , Quinasa de Cadena Ligera de Miosina/deficiencia
20.
Am J Pathol ; 170(2): 439-46, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17255312

RESUMEN

This study investigated the consequences of deletion of the long isoform of myosin light chain kinase (MLCK210) on the cardiovascular changes induced by the bacterial endotoxin lipopolysaccharide (LPS) and cecal ligation puncture using MLCK210-/- mice. Here, we provide evidence that deletion of MLCK210 enhanced survival after intraperitoneal injection of LPS or cecal ligation puncture. LPS-induced vascular hyporeactivity to vasoconstrictor agents was completely prevented in aorta from MLCK210-/- mice. This was associated with a decreased up-regulation of nuclear facor-kappaB expression and activity, inducible nitric-oxide synthase, and level of oxidative stress in the vascular media. Furthermore, LPS-induced increase of nitric oxide production in the circulation and tissues (including heart, liver, and lung) that was correlated with an increased expression of inducible nitric-oxide synthase was also reduced in MLCK210-/- mice. These data demonstrate a role for MLCK210 in endotoxin shock injury associated with oxidative and nitrosative stresses and vascular hyporeactivity.


Asunto(s)
Quinasa de Cadena Ligera de Miosina/metabolismo , Estrés Oxidativo , Choque Séptico/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Eliminación de Gen , Lipopolisacáridos/toxicidad , Hígado/metabolismo , Hígado/patología , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Noqueados , Miocardio/metabolismo , Miocardio/patología , Quinasa de Cadena Ligera de Miosina/deficiencia , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Choque Séptico/inducido químicamente , Choque Séptico/genética , Choque Séptico/patología , Choque Séptico/prevención & control , Regulación hacia Arriba/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Vasoconstricción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA