Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Physiol Heart Circ Physiol ; 326(1): H180-H189, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-37999644

RESUMEN

During select pathological conditions, the heart can hypertrophy and remodel in either a dilated or concentric ventricular geometry, which is associated with lengthening or widening of cardiomyocytes, respectively. The mitogen-activated protein kinase kinase 1 (MEK1) and extracellular signal-related kinase 1 and 2 (ERK1/2) pathway has been implicated in these differential types of growth such that cardiac overexpression of activated MEK1 causes profound concentric hypertrophy and cardiomyocyte thickening, while genetic ablation of the genes encoding ERK1/2 in the mouse heart causes dilation and cardiomyocyte lengthening. However, the mechanisms by which this kinase signaling pathway controls cardiomyocyte directional growth as well as its downstream effectors are poorly understood. To investigate this, we conducted an unbiased phosphoproteomic screen in cultured neonatal rat ventricular myocytes treated with an activated MEK1 adenovirus, the MEK1 inhibitor U0126, or an eGFP adenovirus control. Bioinformatic analysis identified cytoskeletal-related proteins as the largest subset of differentially phosphorylated proteins. Phos-tag and traditional Western blotting were performed to confirm that many cytoskeletal proteins displayed changes in phosphorylation with manipulations in MEK1-ERK1/2 signaling. From this, we hypothesized that the actin cytoskeleton would be changed in vivo in the mouse heart. Indeed, we found that activated MEK1 transgenic mice and gene-deleted mice lacking ERK1/2 protein had enhanced non-sarcomeric actin expression in cardiomyocytes compared with wild-type control hearts. Consistent with these results, cytoplasmic ß- and γ-actin were increased at the subcortical intracellular regions of adult cardiomyocytes. Together, these data suggest that MEK1-ERK1/2 signaling influences the non-sarcomeric cytoskeletal actin network, which may be important for facilitating the growth of cardiomyocytes in length and/or width.NEW & NOTEWORTHY Here, we performed an unbiased analysis of the total phosphoproteome downstream of MEK1-ERK1/2 kinase signaling in cardiomyocytes. Pathway analysis suggested that proteins of the non-sarcomeric cytoskeleton were the most differentially affected. We showed that cytoplasmic ß-actin and γ-actin isoforms, regulated by MEK1-ERK1/2, are localized to the subcortical space at both lateral membranes and intercalated discs of adult cardiomyocytes suggesting how MEK1-ERK1/2 signaling might underlie directional growth of adult cardiomyocytes.


Asunto(s)
Actinas , Miocitos Cardíacos , Ratones , Ratas , Animales , Miocitos Cardíacos/metabolismo , Actinas/metabolismo , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Transducción de Señal , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Citoesqueleto/metabolismo , Ratones Transgénicos , Hipertrofia/metabolismo , Hipertrofia/patología , Proteínas del Citoesqueleto/metabolismo , Células Cultivadas
2.
Nature ; 577(7790): 405-409, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31775156

RESUMEN

Clinical trials using adult stem cells to regenerate damaged heart tissue continue to this day1,2, despite ongoing questions of efficacy and a lack of mechanistic understanding of the underlying biological effect3. The rationale for these cell therapy trials is derived from animal studies that show a modest but reproducible improvement in cardiac function in models of cardiac ischaemic injury4,5. Here we examine the mechanistic basis for cell therapy in mice after ischaemia-reperfusion injury, and find that-although heart function is enhanced-it is not associated with the production of new cardiomyocytes. Cell therapy improved heart function through an acute sterile immune response characterized by the temporal and regional induction of CCR2+ and CX3CR1+ macrophages. Intracardiac injection of two distinct types of adult stem cells, cells killed by freezing and thawing or a chemical inducer of the innate immune response all induced a similar regional accumulation of CCR2+ and CX3CR1+ macrophages, and provided functional rejuvenation to the heart after ischaemia-reperfusion injury. This selective macrophage response altered the activity of cardiac fibroblasts, reduced the extracellular matrix content in the border zone and enhanced the mechanical properties of the injured area. The functional benefit of cardiac cell therapy is thus due to an acute inflammatory-based wound-healing response that rejuvenates the infarcted area of the heart.


Asunto(s)
Inmunidad Innata , Miocitos Cardíacos/inmunología , Trasplante de Células Madre , Células Madre , Animales , Diferenciación Celular , Femenino , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/trasplante , Rejuvenecimiento
3.
Nat Commun ; 10(1): 76, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30622267

RESUMEN

Thrombospondins (Thbs) are a family of five secreted matricellular glycoproteins in vertebrates that broadly affect cell-matrix interaction. While Thbs4 is known to protect striated muscle from disease by enhancing sarcolemmal stability through increased integrin and dystroglycan attachment complexes, here we show that Thbs3 antithetically promotes sarcolemmal destabilization by reducing integrin function, augmenting disease-induced decompensation. Deletion of Thbs3 in mice enhances integrin membrane expression and membrane stability, protecting the heart from disease stimuli. Transgene-mediated overexpression of α7ß1D integrin in the heart ameliorates the disease predisposing effects of Thbs3 by augmenting sarcolemmal stability. Mechanistically, we show that mutating Thbs3 to contain the conserved RGD integrin binding domain normally found in Thbs4 and Thbs5 now rescues the defective expression of integrins on the sarcolemma. Thus, Thbs proteins mediate the intracellular processing of integrin plasma membrane attachment complexes to regulate the dynamics of cellular remodeling and membrane stability.


Asunto(s)
Cardiomiopatías/patología , Integrinas/metabolismo , Sarcolema/patología , Trombospondinas/metabolismo , Animales , Células COS , Cardiomiopatías/diagnóstico por imagen , Cardiomiopatías/etiología , Células Cultivadas , Chlorocebus aethiops , Modelos Animales de Enfermedad , Distroglicanos/metabolismo , Ecocardiografía , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Miocitos Cardíacos , Cultivo Primario de Células , Dominios y Motivos de Interacción de Proteínas/genética , Ratas , Ratas Sprague-Dawley , Sarcolema/metabolismo , Trombospondinas/genética
4.
Circ Genom Precis Med ; 11(3): e001901, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29540468

RESUMEN

BACKGROUND: Inhibition of PKC-α (protein kinase C-α) enhances contractility and cardioprotection in animal models, but effects in humans are unknown. Genotypes at rs9912468 strongly associate with PRKCA expression in the left ventricle, enabling genetic approaches to measure effects of reduced PKC-α in human populations. METHODS AND RESULTS: We analyzed the cis expression quantitative trait locus for PRKCA marked by rs9912468 using 313 left ventricular specimens from European Ancestry patients. The forward strand minor allele (G) at rs9912468 is associated with reduced PKC-α transcript abundance (1.7-fold reduction in minor allele homozygotes, P=1×10-41). This association was cardiac specific in expression quantitative trait locus data sets that span 16 human tissues. Cardiac epigenomic data revealed a predicted enhancer in complete (R2=1.0) linkage disequilibrium with rs9912468 within intron 2 of PRKCA. We cloned this region and used reporter constructs to verify cardiac-specific enhancer activity in vitro in cardiac and noncardiac cells and in vivo in zebrafish. The PRKCA enhancer contains 2 common genetic variants and 4 haplotypes; the haplotype correlated with the rs9912468 PKC-α-lowering allele (G) showed lowest activity. In contrast to previous reports in animal models, the PKC-α-lowering allele is associated with adverse left ventricular remodeling (higher mass, larger diastolic dimension), reduced fractional shortening, and higher risk of dilated cardiomyopathy in human populations. CONCLUSIONS: These findings support PKC-α as a regulator of the human heart but suggest that PKC-α inhibition may adversely affect the left ventricle depending on timing and duration. Pharmacological studies in human subjects are required to discern potential benefits and harms of PKC-α inhibitors as an approach to treat heart disease.


Asunto(s)
Ventrículos Cardíacos/metabolismo , Proteína Quinasa C-alfa/genética , Remodelación Ventricular/genética , Adulto , Anciano , Alelos , Animales , Femenino , Genes Reporteros , Predisposición Genética a la Enfermedad , Genotipo , Haplotipos , Homocigoto , Humanos , Intrones , Desequilibrio de Ligamiento , Masculino , Persona de Mediana Edad , Proteína Quinasa C-alfa/metabolismo , Sitios de Carácter Cuantitativo , Pez Cebra
6.
7.
Circ Res ; 119(2): 249-60, 2016 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-27225478

RESUMEN

RATIONALE: Mitogen-activated protein kinase (MAPK) signaling regulates the growth response of the adult myocardium in response to increased cardiac workload or pathological insults. The dual-specificity phosphatases (DUSPs) are critical effectors, which dephosphorylate the MAPKs to control the basal tone, amplitude, and duration of MAPK signaling. OBJECTIVE: To examine DUSP8 as a regulator of MAPK signaling in the heart and its impact on ventricular and cardiac myocyte growth dynamics. METHODS AND RESULTS: Dusp8 gene-deleted mice and transgenic mice with inducible expression of DUSP8 in the heart were used here to investigate how this MAPK-phosphatase might regulate intracellular signaling and cardiac growth dynamics in vivo. Dusp8 gene-deleted mice were mildly hypercontractile at baseline with a cardiac phenotype of concentric ventricular remodeling, which protected them from progressing towards heart failure in 2 surgery-induced disease models. Cardiac-specific overexpression of DUSP8 produced spontaneous eccentric remodeling and ventricular dilation with heart failure. At the cellular level, adult cardiac myocytes from Dusp8 gene-deleted mice were thicker and shorter, whereas DUSP8 overexpression promoted cardiac myocyte lengthening with a loss of thickness. Mechanistically, activation of extracellular signal-regulated kinases 1/2 were selectively increased in Dusp8 gene-deleted hearts at baseline and following acute pathological stress stimulation, whereas p38 MAPK and c-Jun N-terminal kinases were mostly unaffected. CONCLUSIONS: These results indicate that DUSP8 controls basal and acute stress-induced extracellular signal-regulated kinases 1/2 signaling in adult cardiac myocytes that then alters the length-width growth dynamics of individual cardiac myocytes, which further alters contractility, ventricular remodeling, and disease susceptibility.


Asunto(s)
Fosfatasas de Especificidad Dual/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Miocitos Cardíacos/fisiología , Remodelación Ventricular/fisiología , Animales , Animales Recién Nacidos , Células Cultivadas , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ratas
8.
J Biol Chem ; 291(19): 9920-8, 2016 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-26966179

RESUMEN

Duchenne muscular dystrophy (DMD) is an X-linked recessive disease caused by mutations in the gene encoding dystrophin. Loss of dystrophin protein compromises the stability of the sarcolemma membrane surrounding each muscle cell fiber, leading to membrane ruptures and leakiness that induces myofiber necrosis, a subsequent inflammatory response, and progressive tissue fibrosis with loss of functional capacity. Cathepsin S (Ctss) is a cysteine protease that is actively secreted in areas of tissue injury and ongoing inflammation, where it participates in extracellular matrix remodeling and healing. Here we show significant induction of Ctss expression and proteolytic activity following acute muscle injury or in muscle from mdx mice, a model of DMD. To examine the functional ramifications associated with greater Ctss expression, the Ctss gene was deleted in the mdx genetic background, resulting in protection from muscular dystrophy pathogenesis that included reduced myofiber turnover and histopathology, reduced fibrosis, and improved running capacity. Mechanistically, deletion of the Ctss gene in the mdx background significantly increased myofiber sarcolemmal membrane stability with greater expression and membrane localization of utrophin, integrins, and ß-dystroglycan, which anchor the membrane to the basal lamina and underlying cytoskeletal proteins. Consistent with these results, skeletal muscle-specific transgenic mice overexpressing Ctss showed increased myofiber necrosis, muscle histopathology, and a functional deficit reminiscent of muscular dystrophy. Hence, Ctss induction during muscular dystrophy is a pathologic event that partially underlies disease pathogenesis, and its inhibition might serve as a new therapeutic strategy in DMD.


Asunto(s)
Catepsinas/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Fibras Musculares Esqueléticas/enzimología , Distrofia Muscular Animal/enzimología , Distrofia Muscular de Duchenne/enzimología , Animales , Citoesqueleto/enzimología , Citoesqueleto/genética , Citoesqueleto/patología , Ratones , Ratones Endogámicos mdx , Ratones Noqueados , Fibras Musculares Esqueléticas/patología , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Necrosis , Proteolisis , Sarcolema/enzimología , Sarcolema/genética , Sarcolema/patología
9.
Nature ; 509(7500): 337-41, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24805242

RESUMEN

If and how the heart regenerates after an injury event is highly debated. c-kit-expressing cardiac progenitor cells have been reported as the primary source for generation of new myocardium after injury. Here we generated two genetic approaches in mice to examine whether endogenous c-kit(+) cells contribute differentiated cardiomyocytes to the heart during development, with ageing or after injury in adulthood. A complementary DNA encoding either Cre recombinase or a tamoxifen-inducible MerCreMer chimaeric protein was targeted to the Kit locus in mice and then bred with reporter lines to permanently mark cell lineage. Endogenous c-kit(+) cells did produce new cardiomyocytes within the heart, although at a percentage of approximately 0.03 or less, and if a preponderance towards cellular fusion is considered, the percentage falls to below approximately 0.008. By contrast, c-kit(+) cells amply generated cardiac endothelial cells. Thus, endogenous c-kit(+) cells can generate cardiomyocytes within the heart, although probably at a functionally insignificant level.


Asunto(s)
Linaje de la Célula , Lesiones Cardíacas/patología , Mioblastos Cardíacos/citología , Mioblastos Cardíacos/metabolismo , Miocardio/citología , Miocitos Cardíacos/citología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Envejecimiento/fisiología , Animales , Diferenciación Celular , Fusión Celular , Células Endoteliales/citología , Células Endoteliales/metabolismo , Femenino , Corazón/crecimiento & desarrollo , Integrasas/genética , Integrasas/metabolismo , Masculino , Ratones , Modelos Biológicos , Miocitos Cardíacos/metabolismo , Regeneración/fisiología , Tamoxifeno/farmacología
10.
J Clin Invest ; 123(1): 37-45, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23281408

RESUMEN

Cardiovascular disease is the number one cause of mortality in the Western world. The heart responds to many cardiopathological conditions with hypertrophic growth by enlarging individual myocytes to augment cardiac pump function and decrease ventricular wall tension. Initially, such cardiac hypertrophic growth is often compensatory, but as time progresses these changes become maladaptive. Cardiac hypertrophy is the strongest predictor for the development of heart failure, arrhythmia, and sudden death. Here we discuss therapeutic avenues emerging from molecular and genetic studies of cardiovascular disease in animal models. The majority of these are based on intracellular signaling pathways considered central to pathologic cardiac remodeling and hypertrophy, which then leads to heart failure. We focus our discussion on selected therapeutic targets that have more recently emerged and have a tangible translational potential given the available pharmacologic agents that could be readily evaluated in human clinical trials.


Asunto(s)
Cardiomegalia/tratamiento farmacológico , Cardiomegalia/metabolismo , Transducción de Señal , Remodelación Ventricular , Animales , Arritmias Cardíacas/tratamiento farmacológico , Arritmias Cardíacas/etiología , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/mortalidad , Arritmias Cardíacas/patología , Arritmias Cardíacas/fisiopatología , Cardiomegalia/complicaciones , Cardiomegalia/mortalidad , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Ensayos Clínicos como Asunto , Muerte Súbita/epidemiología , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/mortalidad , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Humanos
11.
Nat Rev Mol Cell Biol ; 14(1): 38-48, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23258295

RESUMEN

The heart hypertrophies in response to developmental signals as well as increased workload. Although adult-onset hypertrophy can ultimately lead to disease, cardiac hypertrophy is not necessarily maladaptive and can even be beneficial. Progress has been made in our understanding of the structural and molecular characteristics of physiological cardiac hypertrophy, as well as of the endocrine effectors and associated signalling pathways that regulate it. Physiological hypertrophy is initiated by finite signals, which include growth hormones (such as thyroid hormone, insulin, insulin-like growth factor 1 and vascular endothelial growth factor) and mechanical forces that converge on a limited number of intracellular signalling pathways (such as PI3K, AKT, AMP-activated protein kinase and mTOR) to affect gene transcription, protein translation and metabolism. Harnessing adaptive signalling mediators to reinvigorate the diseased heart could have important medical ramifications.


Asunto(s)
Cardiomegalia/metabolismo , Corazón/fisiología , Animales , Metabolismo Energético , Insuficiencia Cardíaca/metabolismo , Humanos , Ratones , Biosíntesis de Proteínas , Transducción de Señal , Hormonas Tiroideas/metabolismo , Transcripción Genética
12.
J Cell Biol ; 199(5): 783-98, 2012 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-23166348

RESUMEN

Inositol 1,4,5'-triphosphate receptor II (IP(3)RII) calcium channel expression is increased in both hypertrophic failing human myocardium and experimentally induced models of the disease. The ectopic calcium released from these receptors induces pro-hypertrophic gene expression and may promote arrhythmias. Here, we show that IP(3)RII expression was constitutively restrained by the muscle-specific miRNA, miR-133a. During the hypertrophic response to pressure overload or neurohormonal stimuli, miR-133a down-regulation permitted IP(3)RII levels to increase, instigating pro-hypertrophic calcium signaling and concomitant pathological remodeling. Using a combination of in vivo and in vitro approaches, we demonstrated that IP(3)-induced calcium release (IICR) initiated the hypertrophy-associated decrease in miR-133a. In this manner, hypertrophic stimuli that engage IICR set a feed-forward mechanism in motion whereby IICR decreased miR-133a expression, further augmenting IP(3)RII levels and therefore pro-hypertrophic calcium release. Consequently, IICR can be considered as both an initiating event and a driving force for pathological remodeling.


Asunto(s)
Señalización del Calcio , Cardiomegalia/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , MicroARNs/metabolismo , Animales , Células Cultivadas , Células HEK293 , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , MicroARNs/genética , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Ratas Wistar
13.
J Cell Biol ; 198(5): 815-32, 2012 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-22945933

RESUMEN

Satellite cells (SCs) are stem cells that mediate skeletal muscle growth and regeneration. Here, we observe that adult quiescent SCs and their activated descendants expressed the homeodomain transcription factor Six1. Genetic disruption of Six1 specifically in adult SCs impaired myogenic cell differentiation, impaired myofiber repair during regeneration, and perturbed homeostasis of the stem cell niche, as indicated by an increase in SC self-renewal. Six1 regulated the expression of the myogenic regulatory factors MyoD and Myogenin, but not Myf5, which suggests that Six1 acts on divergent genetic networks in the embryo and in the adult. Moreover, we demonstrate that Six1 regulates the extracellular signal-regulated kinase 1/2 (ERK1/2) pathway during regeneration via direct control of Dusp6 transcription. Muscles lacking Dusp6 were able to regenerate properly but showed a marked increase in SC number after regeneration. We conclude that Six1 homeoproteins act as a rheostat system to ensure proper regeneration of the tissue and replenishment of the stem cell pool during the events that follow skeletal muscle trauma.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Músculo Esquelético/fisiología , Regeneración/fisiología , Células Satélite del Músculo Esquelético/fisiología , Células Madre/fisiología , Cicatrización de Heridas/fisiología , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Fosfatasa 6 de Especificidad Dual/genética , Fosfatasa 6 de Especificidad Dual/metabolismo , Proteínas de Homeodominio/genética , Homeostasis , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Proteína MioD/genética , Proteína MioD/metabolismo , Miofibrillas/metabolismo , Miofibrillas/fisiología , Factor 5 Regulador Miogénico/genética , Factor 5 Regulador Miogénico/metabolismo , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Miogenina/genética , Miogenina/metabolismo , Regeneración/genética , Células Satélite del Músculo Esquelético/metabolismo , Células Madre/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Cicatrización de Heridas/genética
14.
Circ Res ; 111(6): 761-77, 2012 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-22935533

RESUMEN

The advent of modern mouse genetics has benefited many fields of diseased-based research over the past 20 years, none perhaps more profoundly than cardiac biology. Indeed, the heart is now arguably one of the easiest tissues to genetically manipulate, given the availability of an ever-growing tool chest of molecular reagents/promoters and "facilitator" mouse lines. It is now possible to modify the expression of essentially any gene or partial gene product in the mouse heart at any time, either gain or loss of function. This review is designed as a handbook for the nonmouse geneticist and/or junior investigator to permit the successful manipulation of any gene or RNA product in the heart, while avoiding artifacts. In the present review, guidelines, pitfalls, and limitations are presented so that rigorous and appropriate examination of cardiac genotype-phenotype relationships can be performed. This review uses examples from the field to illustrate the vast spectrum of experimental and design details that must be considered when using genetically modified mouse models to study cardiac biology.


Asunto(s)
Investigación Biomédica/métodos , Modelos Animales de Enfermedad , Corazón/fisiopatología , Miocardio/metabolismo , Animales , Guías como Asunto , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Miocardio/patología
15.
Cell ; 149(6): 1257-68, 2012 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-22682248

RESUMEN

Thrombospondin (Thbs) proteins are induced in sites of tissue damage or active remodeling. The endoplasmic reticulum (ER) stress response is also prominently induced with disease where it regulates protein production and resolution of misfolded proteins. Here we describe a function for Thbs as ER-resident effectors of an adaptive ER stress response. Thbs4 cardiac-specific transgenic mice were protected from myocardial injury, whereas Thbs4(-/-) mice were sensitized to cardiac maladaptation. Thbs induction produced a unique profile of adaptive ER stress response factors and expansion of the ER and downstream vesicles. Thbs bind the ER lumenal domain of activating transcription factor 6α (Atf6α) to promote its nuclear shuttling. Thbs4(-/-) mice showed blunted activation of Atf6α and other ER stress-response factors with injury, and Thbs4-mediated protection was lost upon Atf6α deletion. Hence, Thbs can function inside the cell during disease remodeling to augment ER function and protect through a mechanism involving regulation of Atf6α.


Asunto(s)
Estrés del Retículo Endoplásmico , Transducción de Señal , Trombospondinas/metabolismo , Factor de Transcripción Activador 6/genética , Animales , Cardiomiopatías/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Trombospondinas/genética
16.
J Cell Biol ; 193(7): 1181-96, 2011 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-21690310

RESUMEN

Unraveling the gene regulatory networks that govern development and function of the mammalian heart is critical for the rational design of therapeutic interventions in human heart disease. Using the Drosophila heart as a platform for identifying novel gene interactions leading to heart disease, we found that the Rho-GTPase Cdc42 cooperates with the cardiac transcription factor Tinman/Nkx2-5. Compound Cdc42, tinman heterozygous mutant flies exhibited impaired cardiac output and altered myofibrillar architecture, and adult heart-specific interference with Cdc42 function is sufficient to cause these same defects. We also identified K(+) channels, encoded by dSUR and slowpoke, as potential effectors of the Cdc42-Tinman interaction. To determine whether a Cdc42-Nkx2-5 interaction is conserved in the mammalian heart, we examined compound heterozygous mutant mice and found conduction system and cardiac output defects. In exploring the mechanism of Nkx2-5 interaction with Cdc42, we demonstrated that mouse Cdc42 was a target of, and negatively regulated by miR-1, which itself was negatively regulated by Nkx2-5 in the mouse heart and by Tinman in the fly heart. We conclude that Cdc42 plays a conserved role in regulating heart function and is an indirect target of Tinman/Nkx2-5 via miR-1.


Asunto(s)
Proteínas de Drosophila/fisiología , Drosophila/fisiología , Corazón/fisiología , Proteínas de Homeodominio/fisiología , MicroARNs/fisiología , Proteínas Represoras/fisiología , Transactivadores/fisiología , Factores de Transcripción/fisiología , Proteína de Unión al GTP cdc42/metabolismo , Animales , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Proteínas de Unión al GTP/fisiología , Regulación del Desarrollo de la Expresión Génica , Cardiopatías/genética , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/genética , Humanos , Ratones , Contracción Miocárdica/genética , Miocardio/metabolismo , Miocitos Cardíacos/citología , Proteínas Represoras/genética , Transactivadores/genética , Factores de Transcripción/genética , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/fisiología , Proteínas de Unión al GTP rho/metabolismo , Proteínas de Unión al GTP rho/fisiología
17.
Circ Res ; 109(2): 141-50, 2011 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-21597010

RESUMEN

RATIONALE: Cardiac myosin-binding protein-C (cMyBP-C) phosphorylation at Ser-273, Ser-282, and Ser-302 regulates myocardial contractility. In vitro and in vivo experiments suggest the nonequivalence of these sites and the potential importance of Ser-282 phosphorylation in modulating the protein's overall phosphorylation and myocardial function. OBJECTIVE: To determine whether complete cMyBP-C phosphorylation is dependent on Ser-282 phosphorylation and to define its role in myocardial function. We hypothesized that Ser-282 regulates Ser-302 phosphorylation and cardiac function during ß-adrenergic stimulation. METHODS AND RESULTS: Using recombinant human C1-M-C2 peptides in vitro, we determined that protein kinase A can phosphorylate Ser-273, Ser-282, and Ser-302. Protein kinase C can also phosphorylate Ser-273 and Ser-302. In contrast, Ca(2+)-calmodulin-activated kinase II targets Ser-302 but can also target Ser-282 at nonphysiological calcium concentrations. Strikingly, Ser-302 phosphorylation by Ca(2+)-calmodulin-activated kinase II was abolished by ablating the ability of Ser-282 to be phosphorylated via alanine substitution. To determine the functional roles of the sites in vivo, three transgenic lines, which expressed cMyBP-C containing either Ser-273-Ala-282-Ser-302 (cMyBP-C(SAS)), Ala-273-Asp-282-Ala-302 (cMyBP-C(ADA)), or Asp-273-Ala-282-Asp-302 (cMyBP-C(DAD)), were generated. Mutant protein was completely substituted for endogenous cMyBP-C by breeding each mouse line into a cMyBP-C null (t/t) background. Serine-to-alanine substitutions were used to ablate the abilities of the residues to be phosphorylated, whereas serine-to-aspartate substitutions were used to mimic the charged state conferred by phosphorylation. Compared to control nontransgenic mice, as well as transgenic mice expressing wild-type cMyBP-C, the transgenic cMyBP-C(SAS(t/t)), cMyBP-C(ADA(t/t)), and cMyBP-C(DAD(t/t)) mice showed no increases in morbidity and mortality and partially rescued the cMyBP-C((t/t)) phenotype. The loss of cMyBP-C phosphorylation at Ser-282 led to an altered ß-adrenergic response. In vivo hemodynamic studies revealed that contractility was unaffected but that cMyBP-C(SAS(t/t)) hearts showed decreased diastolic function at baseline. However, the normal increases in cardiac function (increased contractility/relaxation) as a result of infusion of ß-agonist was significantly decreased in all of the mutants, suggesting that competency for phosphorylation at multiple sites in cMyBP-C is a prerequisite for normal ß-adrenergic responsiveness. CONCLUSIONS: Ser-282 has a unique regulatory role in that its phosphorylation is critical for the subsequent phosphorylation of Ser-302. However, each residue plays a role in regulating the contractile response to ß-agonist stimulation.


Asunto(s)
Proteínas Portadoras/metabolismo , Corazón/fisiología , Serina/metabolismo , Agonistas Adrenérgicos beta/farmacología , Sustitución de Aminoácidos , Animales , Proteínas Portadoras/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Humanos , Ratones , Ratones Transgénicos , Contracción Miocárdica/efectos de los fármacos , Fosforilación
18.
Circ Res ; 108(2): 176-83, 2011 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-21127295

RESUMEN

RATIONALE: An increase in cardiac afterload typically produces concentric hypertrophy characterized by an increase in cardiomyocyte width, whereas volume overload or exercise results in eccentric growth characterized by cellular elongation and addition of sarcomeres in series. The signaling pathways that control eccentric versus concentric heart growth are not well understood. OBJECTIVE: To determine the role of extracellular signal-regulated kinase 1 and 2 (ERK1/2) in regulating the cardiac hypertrophic response. METHODS AND RESULTS: Here, we used mice lacking all ERK1/2 protein in the heart (Erk1(-/-) Erk2(fl/fl-Cre)) and mice expressing activated mitogen-activated protein kinase kinase (Mek)1 in the heart to induce ERK1/2 signaling, as well as mechanistic experiments in cultured myocytes to assess cellular growth characteristics associated with this signaling pathway. Although genetic deletion of all ERK1/2 from the mouse heart did not block the cardiac hypertrophic response per se, meaning that the heart still increased in weight with both aging and pathological stress stimulation, it did dramatically alter how the heart grew. For example, adult myocytes from hearts of Erk1(-/-) Erk2(fl/fl-Cre) mice showed preferential eccentric growth (lengthening), whereas myocytes from Mek1 transgenic hearts showed concentric growth (width increase). Isolated adult myocytes acutely inhibited for ERK1/2 signaling by adenoviral gene transfer showed spontaneous lengthening, whereas infection with an activated Mek1 adenovirus promoted constitutive ERK1/2 signaling and increased myocyte thickness. A similar effect was observed in engineered heart tissue under cyclic stretching, where ERK1/2 inhibition led to preferential lengthening. CONCLUSIONS: Taken together, these data demonstrate that the ERK1/2 signaling pathway uniquely regulates the balance between eccentric and concentric growth of the heart.


Asunto(s)
Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Animales , Células Cultivadas , Hipertrofia , MAP Quinasa Quinasa 1/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Modelos Animales , Fosforilación , Transducción de Señal/fisiología
19.
Circ Res ; 107(5): 659-66, 2010 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-20616315

RESUMEN

RATIONALE: Inositol 1,4,5-trisphosphate (IP(3)) is a second messenger that regulates intracellular Ca(2+) release through IP(3) receptors located in the sarco(endo)plasmic reticulum of cardiac myocytes. Many prohypertrophic G protein-coupled receptor (GPCR) signaling events lead to IP(3) liberation, although its importance in transducing the hypertrophic response has not been established in vivo. OBJECTIVE: Here, we generated conditional, heart-specific transgenic mice with both gain- and loss-of-function for IP(3) receptor signaling to examine its hypertrophic growth effects following pathological and physiological stimulation. METHODS AND RESULTS: Overexpression of the mouse type-2 IP(3) receptor (IP(3)R2) in the heart generated mild baseline cardiac hypertrophy at 3 months of age. Isolated myocytes from overexpressing lines showed increased Ca(2+) transients and arrhythmias in response to endothelin-1 stimulation. Although low levels of IP(3)R2 overexpression failed to augment/synergize cardiac hypertrophy following 2 weeks of pressure-overload stimulation, such levels did enhance hypertrophy following 2 weeks of isoproterenol infusion, in response to Galphaq overexpression, and/or in response to exercise stimulation. To inhibit IP(3) signaling in vivo, we generated transgenic mice expressing an IP(3) chelating protein (IP(3)-sponge). IP(3)-sponge transgenic mice abrogated cardiac hypertrophy in response to isoproterenol and angiotensin II infusion but not pressure-overload stimulation. Mechanistically, IP(3)R2-enhanced cardiac hypertrophy following isoproterenol infusion was significantly reduced in the calcineurin-Abeta-null background. CONCLUSION: These results indicate that IP(3)-mediated Ca(2+) release plays a central role in regulating cardiac hypertrophy downstream of GPCR signaling, in part, through a calcineurin-dependent mechanism.


Asunto(s)
Señalización del Calcio , Cardiomegalia/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Inositol 1,4,5-Trifosfato/metabolismo , Miocitos Cardíacos/metabolismo , Factores de Edad , Angiotensina II , Animales , Arritmias Cardíacas/inducido químicamente , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Calcineurina/deficiencia , Calcineurina/genética , Cardiomegalia/inducido químicamente , Cardiomegalia/genética , Cardiomegalia/patología , Cardiomegalia/prevención & control , Modelos Animales de Enfermedad , Endotelina-1 , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/deficiencia , Receptores de Inositol 1,4,5-Trifosfato/genética , Isoproterenol , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Miocitos Cardíacos/patología , Fenotipo , Esfuerzo Físico
20.
J Biol Chem ; 285(9): 6716-24, 2010 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-20037164

RESUMEN

Calcineurin is a protein phosphatase that is uniquely regulated by sustained increases in intracellular Ca(2+) following signal transduction events. Calcineurin controls cellular proliferation, differentiation, apoptosis, and inducible gene expression following stress and neuroendocrine stimulation. In the adult heart, calcineurin regulates hypertrophic growth of cardiomyocytes in response to pathologic insults that are associated with altered Ca(2+) handling. Here we determined that calcineurin signaling is directly linked to the proper control of cardiac contractility, rhythm, and the expression of Ca(2+)-handling genes in the heart. Our approach involved a cardiomyocyte-specific deletion using a CnB1-LoxP-targeted allele in mice and three different cardiac-expressing Cre alleles/transgenes. Deletion of calcineurin with the Nkx2.5-Cre knock-in allele resulted in lethality at 1 day after birth due to altered right ventricular morphogenesis, reduced ventricular trabeculation, septal defects, and valvular overgrowth. Slightly later deletion of calcineurin with the alpha-myosin heavy chain Cre transgene resulted in lethality in early mid adulthood that was characterized by substantial reductions in cardiac contractility, severe arrhythmia, and reduced myocyte content in the heart. Young calcineurin heart-deleted mice died suddenly after pressure overload stimulation or neuroendocrine agonist infusion, and telemetric monitoring of older mice showed arrhythmia leading to sudden death. Mechanistically, loss of calcineurin reduced expression of key Ca(2+)-handling genes that likely lead to arrhythmia and reduced contractility. Loss of calcineurin also directly impacted cellular proliferation in the postnatal developing heart. These results reveal multiple mechanisms whereby calcineurin regulates cardiac development and myocyte contractility.


Asunto(s)
Calcineurina/fisiología , Corazón/crecimiento & desarrollo , Animales , Arritmias Cardíacas , Calcineurina/deficiencia , Calcineurina/genética , Calcio , Proliferación Celular , Eliminación de Gen , Corazón/fisiología , Ratones , Ratones Transgénicos , Contracción Miocárdica , Subunidades de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...