Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 54(9): 2072-2088.e7, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34320366

RESUMEN

Cardiac macrophages represent a heterogeneous cell population with distinct origins, dynamics, and functions. Recent studies have revealed that C-C Chemokine Receptor 2 positive (CCR2+) macrophages derived from infiltrating monocytes regulate myocardial inflammation and heart failure pathogenesis. Comparatively little is known about the functions of tissue resident (CCR2-) macrophages. Herein, we identified an essential role for CCR2- macrophages in the chronically failing heart. Depletion of CCR2- macrophages in mice with dilated cardiomyopathy accelerated mortality and impaired ventricular remodeling and coronary angiogenesis, adaptive changes necessary to maintain cardiac output in the setting of reduced cardiac contractility. Mechanistically, CCR2- macrophages interacted with neighboring cardiomyocytes via focal adhesion complexes and were activated in response to mechanical stretch through a transient receptor potential vanilloid 4 (TRPV4)-dependent pathway that controlled growth factor expression. These findings establish a role for tissue-resident macrophages in adaptive cardiac remodeling and implicate mechanical sensing in cardiac macrophage activation.


Asunto(s)
Cardiomiopatía Dilatada/metabolismo , Activación de Macrófagos/fisiología , Macrófagos/metabolismo , Remodelación Ventricular/fisiología , Animales , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación , Miocardio/metabolismo , Troponina T/genética
2.
Circ Res ; 130(2): 234-248, 2022 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-34875852

RESUMEN

BACKGROUND: During the development of heart failure, a fetal cardiac gene program is reactivated and accelerates pathological cardiac remodeling. We previously reported that a transcriptional repressor, NRSF (neuron restrictive silencer factor), suppresses the fetal cardiac gene program, thereby maintaining cardiac integrity. The underlying molecular mechanisms remain to be determined, however. METHODS: We aim to elucidate molecular mechanisms by which NRSF maintains normal cardiac function. We generated cardiac-specific NRSF knockout mice and analyzed cardiac gene expression profiles in those mice and mice cardiac-specifically expressing a dominant-negative NRSF mutant. RESULTS: We found that cardiac expression of Gαo, an inhibitory G protein encoded in humans by GNAO1, is transcriptionally regulated by NRSF and is increased in the ventricles of several mouse models of heart failure. Genetic knockdown of Gnao1 ameliorated the cardiac dysfunction and prolonged survival rates in these mouse heart failure models. Conversely, cardiac-specific overexpression of GNAO1 in mice was sufficient to induce cardiac dysfunction. Mechanistically, we observed that increasing Gαo expression increased surface sarcolemmal L-type Ca2+ channel activity, activated CaMKII (calcium/calmodulin-dependent kinase-II) signaling, and impaired Ca2+ handling in ventricular myocytes, which led to cardiac dysfunction. CONCLUSIONS: These findings shed light on a novel function of Gαo in the regulation of cardiac Ca2+ homeostasis and systolic function and suggest Gαo may be an effective therapeutic target for the treatment of heart failure.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Insuficiencia Cardíaca/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas Represoras/metabolismo , Animales , Canales de Calcio Tipo L/metabolismo , Señalización del Calcio , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Células Cultivadas , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/metabolismo , Homeostasis , Ratones , Ratones Endogámicos C57BL , Proteínas Represoras/genética
3.
Mol Pharmacol ; 104(6): 275-286, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37678938

RESUMEN

Type 2 ryanodine receptor (RyR2) is a Ca2+ release channel on the endoplasmic (ER)/sarcoplasmic reticulum that plays a central role in the excitation-contraction coupling in the heart. Hyperactivity of RyR2 has been linked to ventricular arrhythmias in patients with catecholaminergic polymorphic ventricular tachycardia and heart failure, where spontaneous Ca2+ release via hyperactivated RyR2 depolarizes diastolic membrane potential to induce triggered activity. In such cases, drugs that suppress RyR2 activity are expected to prevent the arrhythmias, but there is no clinically available RyR2 inhibitors at present. In this study, we searched for RyR2 inhibitors from a well-characterized compound library using a recently developed ER Ca2+-based assay, where the inhibition of RyR2 activity was detected by the increase in ER Ca2+ signals from R-CEPIA1er, a genetically encoded ER Ca2+ indicator, in RyR2-expressing HEK293 cells. By screening 1535 compounds in the library, we identified three compounds (chloroxylenol, methyl orsellinate, and riluzole) that greatly increased the ER Ca2+ signal. All of the three compounds suppressed spontaneous Ca2+ oscillations in RyR2-expressing HEK293 cells and correspondingly reduced the Ca2+-dependent [3H]ryanodine binding activity. In cardiomyocytes from RyR2-mutant mice, the three compounds effectively suppressed abnormal Ca2+ waves without substantial effects on the action-potential-induced Ca2+ transients. These results confirm that ER Ca2+-based screening is useful for identifying modulators of ER Ca2+ release channels and suggest that RyR2 inhibitors have potential to be developed as a new category of antiarrhythmic drugs. SIGNIFICANCE STATEMENT: We successfully identified three compounds having RyR2 inhibitory action from a well-characterized compound library using an endoplasmic reticulum Ca2+-based assay, and demonstrated that these compounds suppressed arrhythmogenic Ca2+ wave generation without substantially affecting physiological action-potential induced Ca2+ transients in cardiomyocytes. This study will facilitate the development of RyR2-specific inhibitors as a potential new class of drugs for life-threatening arrhythmias induced by hyperactivation of RyR2.


Asunto(s)
Miocitos Cardíacos , Canal Liberador de Calcio Receptor de Rianodina , Humanos , Ratones , Animales , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Células HEK293 , Retículo Endoplásmico/metabolismo , Arritmias Cardíacas/metabolismo , Retículo Sarcoplasmático , Señalización del Calcio , Calcio/metabolismo , Mutación
4.
Genes Cells ; 26(8): 583-595, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34060165

RESUMEN

Genetic mutations in actin regulators have been emerging as a cause of cardiomyopathy, although the functional link between actin dynamics and cardiac contraction remains largely unknown. To obtain insight into this issue, we examined the effects of pharmacological inhibition of formins, a major class of actin-assembling proteins. The formin inhibitor SMIFH2 significantly enhanced the cardiac contractility of isolated frog hearts, thereby augmenting cardiac performance. SMIFH2 treatment had no significant effects on the Ca2+ sensitivity of frog muscle fibers. Instead, it unexpectedly increased Ca2+ concentrations of isolated frog cardiomyocytes, suggesting that the inotropic effect is due to enhanced Ca2+ transients. In contrast to frog hearts, the contractility of mouse cardiomyocytes was attenuated by SMIFH2 treatment with decreasing Ca2+ transients. Thus, SMIFH2 has opposing effects on the Ca2+ transient and contractility between frog and mouse cardiomyocytes. We further found that SMIFH2 suppressed Ca2+ -release via type 2 ryanodine receptor (RyR2); this inhibitory effect may explain the species differences, since RyR2 is critical for Ca2+ transients in mouse myocardium but absent in frog myocardium. Although the mechanisms underlying the enhancement of Ca2+ transients in frog cardiomyocytes remain unclear, SMIFH2 differentially affects the cardiac contraction of amphibian and mammalian by differentially modulating their Ca2+ handling.


Asunto(s)
Señalización del Calcio , Corazón/efectos de los fármacos , Contracción Miocárdica , Miocitos Cardíacos/efectos de los fármacos , Animales , Células Cultivadas , Corazón/fisiología , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Rana catesbeiana , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Especificidad de la Especie , Tionas/farmacología , Uracilo/análogos & derivados , Uracilo/farmacología
5.
Pediatr Res ; 89(4): 846-857, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32563186

RESUMEN

BACKGROUND: Dilated cardiomyopathy (DCM) in children is often associated with poor morbidity and mortality and exhibits distinct pathological entities from those of adult DCM. Owing to the limited number of patients and the lack of a good animal model, the molecular mechanisms underlying pediatric DCM remain poorly understood. The purpose of this study is to establish an animal model of neonatal DCM and identify early progression factors. METHODS: Cardiac phenotypes and comprehensive gene expression profiles in homozygous ΔK210 knock-in (TNNT2ΔK210/ΔK210) mice were analyzed and compared to TNNT2+/ΔK210 and wild-type mice at 0 days and 1 week of age. RESULTS: Immediately after birth, the cardiac weight in TNNT2ΔK210/ΔK210 mice was already increased compared to that in TNNT2+/ΔK210 and wild-type mice. Echocardiographic examination of 0-day-old and 1-week-old TNNT2ΔK210/ΔK210 mice revealed similar phenotypes of pediatric DCM. In addition, several genes were significantly upregulated in the ventricular tissues of TNNT2ΔK210/ΔK210 mice, and the KEGG PATHWAY analysis revealed several important pathways such as cancer and focal adhesion that might be associated with the pathogenesis and development of DCM. CONCLUSIONS: TNNT2ΔK210/ΔK210 mice have already developed DCM at birth, indicating that they should be an excellent animal model to identify early progression factors of DCM. IMPACT: TNNT2ΔK210/ΔK210 mice are excellent animal model for DCM. TNNT2ΔK210/ΔK210 mice are excellent animal model to identify early progression factors of DCM. KEGG PATHWAY analysis revealed that several important pathways such as cancer and focal adhesion might be associated with the pathogenesis and development of neonatal DCM.


Asunto(s)
Cardiomiopatía Dilatada/genética , Mutación , Troponina T/genética , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Regulación hacia Abajo , Ecocardiografía , Perfilación de la Expresión Génica , Ventrículos Cardíacos/fisiopatología , Homocigoto , Ratones , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Pronóstico , Regulación hacia Arriba
6.
Am J Physiol Heart Circ Physiol ; 314(6): H1192-H1202, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29451818

RESUMEN

Mutations in genes encoding components of the sarcomere cause cardiomyopathy, which is often associated with abnormal Ca2+ sensitivity of muscle contraction. We have previously shown that a heart-specific myosin light chain phosphatase small subunit (hHS-M21) increases the Ca2+ sensitivity of muscle contraction. The aim of the present study was to investigate the function of hHS-M21 in vivo and the causative role of abnormal Ca2+ sensitivity in cardiomyopathy. We generated transgenic mice with cardiac-specific overexpression of hHS-M21. We confirmed that hHS-M21 increased the Ca2+ sensitivity of cardiac muscle contraction in vivo, which was not followed by an increased phosphorylation of myosin light chain 2 isoforms. hHS-M21 transgenic mice developed severe systolic dysfunction with myocardial fibrosis and degeneration of cardiomyocytes in association with sinus bradycardia and atrioventricular conduction defect. The contractile dysfunction and cardiac fibrosis were improved by treatment with the Rho kinase inhibitor fasudil. Our findings suggested that the overexpression of hHS-M21 results in cardiac dysfunction and conduction disturbance via non-myosin light chain 2 phosphorylation-dependent regulation. NEW & NOTEWORTHY The present study is the first to develop mice with transgenic overexpression of a heart-specific myosin light chain phosphatase small subunit (hHS-M21) and to examine the effects of hHS-M21 on cardiac function. Elevation of hHS-M21 induced heart failure with myocardial fibrosis and degeneration of cardiomyocytes accompanied by supraventricular arrhythmias.


Asunto(s)
Arritmias Cardíacas/enzimología , Insuficiencia Cardíaca/enzimología , Miocitos Cardíacos/enzimología , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Animales , Arritmias Cardíacas/genética , Arritmias Cardíacas/patología , Arritmias Cardíacas/fisiopatología , Señalización del Calcio , Miosinas Cardíacas/metabolismo , Cardiomiopatías/enzimología , Cardiomiopatías/genética , Cardiomiopatías/fisiopatología , Modelos Animales de Enfermedad , Fibrosis , Predisposición Genética a la Enfermedad , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Frecuencia Cardíaca , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Contracción Miocárdica , Miocitos Cardíacos/patología , Cadenas Ligeras de Miosina/metabolismo , Fosfatasa de Miosina de Cadena Ligera/genética , Fenotipo , Fosforilación , Subunidades de Proteína , Regulación hacia Arriba , Disfunción Ventricular Izquierda/enzimología , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/fisiopatología , Función Ventricular Izquierda , Remodelación Ventricular , Quinasas Asociadas a rho/metabolismo
7.
Biochem Biophys Res Commun ; 496(4): 1250-1256, 2018 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-29402414

RESUMEN

AIMS: Cardiac Troponin T (TnT) mutation-linked familial hypertrophic cardiomyopathy (FHC) is known to cause sudden cardiac death at a young age. Here, we investigated the role of the Ca2+ release channel of the cardiac sarcoplasmic reticulum (SR), ryanodine receptor (RyR2), in the pathogenic mechanism of lethal arrhythmia in FHC-related TnT-mutated transgenic mice (TG; TnT-delta160E). METHODS AND RESULTS: In TG cardiomyocytes, the Ca2+ spark frequency (SpF) was much higher than that in non-TG cardiomyocytes. These differences were more pronounced in the presence of isoproterenol (ISO; 10 nM). This increase in SpF was largely reversed by a CaMKII inhibitor (KN-93), but not by a protein kinase A inhibitor (H89). CaMKII phosphorylation at Ser2814 in RyR2 was increased significantly in TG. Spontaneous Ca2+ transients (sCaTs) after cessation of a 1-5 Hz pacing, frequently observed in ISO-treated TG cardiomyocytes, were also attenuated by KN-93, but not by H89. The RyR2 stabilizer dantrolene attenuated Ca2+ sparks and sCaTs in ISO-treated TG cardiomyocytes, indicating that the mutation-linked aberrant Ca2+ release is mediated by destabilized RyR2. CONCLUSIONS: In FHC-linked TnT-mutated hearts, RyR2 is susceptible to CaMKII-mediated phosphorylation, presumably because of a mutation-linked increase in diastolic [Ca2+]i, causing aberrant Ca2+ release leading to lethal arrhythmia.


Asunto(s)
Arritmias Cardíacas/fisiopatología , Señalización del Calcio , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Calcio/metabolismo , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Miocitos Cardíacos/metabolismo , Troponina T/metabolismo , Animales , Arritmias Cardíacas/etiología , Cardiomiopatía Hipertrófica Familiar/complicaciones , Células Cultivadas , Ratones , Ratones Transgénicos , Fosforilación , Retículo Sarcoplasmático/metabolismo
8.
Arch Biochem Biophys ; 637: 40-47, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29169900

RESUMEN

The plant-derived flavonoid, quercetin (QCT), has many biological actions, including cardioprotective actions, resulting from its antioxidant and anti-inflammatory effects. In this study, effects of QCT and its metabolites on the contraction and Ca2+ transients (CaT) of mouse single cardiomyocytes were simultaneously measured and compared with those of isoproterenol and digoxin. Furthermore, cardiac function and plasma concentrations were analyzed after bolus intravenous administration of QCT in mice. QCT and its metabolite, tamarixetin, as well as isoproterenol and digoxin, enhanced the contraction and CaT of cardiomyocytes. The inotropic action of isoproterenol was accompanied by an increase in the velocities of sarcomere shortening and relengthening and CaT decay through activation of cAMP-dependent protein kinase; however, no such lusitropic effects accompanied the inotropic action of QCT, tamarixetin or digoxin. Intravenous administration of QCT to mice resulted in a sustained increase in cardiac systolic function; QCT was rapidly metabolized to tamarixetin and its plasma concentration was maintained at high levels over a similar time frame as the enhancement of cardiac systolic function. These results suggest that QCT exerts a cardiotonic action in vivo at least, in part, through digitalis-like enhancement of CaT by itself and its metabolite tamarixetin.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Cardiotónicos/farmacología , Disacáridos/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Quercetina/análogos & derivados , Quercetina/farmacología , Animales , Cardiotónicos/metabolismo , Glicósidos Digitálicos/farmacología , Digoxina/farmacología , Disacáridos/metabolismo , Técnicas In Vitro , Isoproterenol/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Contracción Miocárdica/efectos de los fármacos , Quercetina/metabolismo
9.
Am J Physiol Heart Circ Physiol ; 310(11): H1808-15, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27106044

RESUMEN

Glycogen synthase kinase-3ß (GSK-3ß) plays a central role in both cardiac physiology and pathology. Herein we want to clarify the role of GSK-3ß in familial dilated cardiomyopathy. We generated a mouse model carrying a heterozygous knockout mutation of GSK-3ß (GSK-3ß(+/-) KO), together with a ΔK210 knockin mutation in cardiac troponin T (ΔK210 cTnT KI), which was proved to be one of the genetic causes of familial dilated cardiomyopathy (DCM). GSK-3ß(+/-) KO prevented the slow and rapid deterioration in left ventricular systolic function accompanying heart failure (HF) in DCM mice with heterozygous and homozygous ΔK210 cTnT KI mutations, respectively. GSK-3ß(+/-) KO also prevented cardiac enlargement, myocardial fibrosis, and cardiomyocyte apoptosis and markedly reduced the expression of cardiac ß-myosin heavy chain isoform, indicative of HF, in DCM mice with homozygous ΔK210 cTnT KI mutation. GSK-3ß(+/-) KO also extended the life span of these DCM mice. This study suggests that the inhibition of GSK-3ß is cardioprotective in familial DCM associated with ΔK210 cTnT mutation.


Asunto(s)
Cardiomiopatía Dilatada/genética , Glucógeno Sintasa Quinasa 3 beta/genética , Miocardio/metabolismo , Troponina T/genética , Disfunción Ventricular Izquierda/genética , Animales , Cardiomiopatía Dilatada/metabolismo , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Ratones , Ratones Transgénicos , Cadenas Pesadas de Miosina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Troponina T/metabolismo , Disfunción Ventricular Izquierda/metabolismo
10.
J Pharmacol Sci ; 127(4): 446-55, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25913757

RESUMEN

Differentiation-inducing factor-1 (DIF-1) produced by Dictyostelium discoideum strongly inhibits the proliferation of various types of cancer cells by suppression of the Wnt/ß-catenin signal transduction pathway. In the present study, we examined the effect of differentiation-inducing factor-3 (DIF-3), a monochlorinated metabolite of DIF-1 that is also produced by D. discoideum, on human colon cancer cell lines HCT-116 and DLD-1. DIF-3 strongly inhibited cell proliferation by arresting the cell cycle at the G0/G1 phase. DIF-3 reduced the expression levels of cyclin D1 and c-Myc by facilitating their degradation via activation of GSK-3ß in a time and dose-dependent manner. In addition, DIF-3 suppressed the expression of T-cell factor 7-like 2, a key transcription factor in the Wnt/ß-catenin signaling pathway, thereby reducing the mRNA levels of cyclin D1 and c-Myc. Subsequently, we examined the in vivo effects of DIF-3 in Mutyh(-/-) mice with oxidative stress-induced intestinal cancers. Repeated oral administration of DIF-3 markedly reduced the number and size of cancers at a level comparable to that of DIF-1. These data suggest that DIF-3 inhibits intestinal cancer cell proliferation in vitro and in vivo, probably by mechanisms similar to those identified in DIF-1 actions, and that DIF-3 may be a potential novel anti-cancer agent.


Asunto(s)
Antineoplásicos , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Hexanonas/farmacología , Administración Oral , Animales , Ciclo Celular/efectos de los fármacos , Proliferación Celular/genética , Ciclina D1/genética , Ciclina D1/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Células HCT116 , Hexanonas/administración & dosificación , Humanos , Ratones Transgénicos , Estrés Oxidativo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/fisiología
11.
J Mol Cell Cardiol ; 63: 69-78, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23863340

RESUMEN

It has been reported that the Frank-Starling mechanism is coordinately regulated in cardiac muscle via thin filament "on-off" equilibrium and titin-based lattice spacing changes. In the present study, we tested the hypothesis that the deletion mutation ΔK210 in the cardiac troponin T gene shifts the equilibrium toward the "off" state and accordingly attenuate the sarcomere length (SL) dependence of active force production, via reduced cross-bridge formation. Confocal imaging in isolated hearts revealed that the cardiomyocytes were enlarged, especially in the longitudinal direction, in ΔK210 hearts, with striation patterns similar to those in wild type (WT) hearts, suggesting that the number of sarcomeres is increased in cardiomyocytes but the sarcomere length remains unaltered. For analysis of the SL dependence of active force, skinned muscle preparations were obtained from the left ventricle of WT and knock-in (ΔK210) mice. An increase in SL from 1.90 to 2.20µm shifted the mid-point (pCa50) of the force-pCa curve leftward by ~0.21pCa units in WT preparations. In ΔK210 muscles, Ca(2+) sensitivity was lower by ~0.37pCa units, and the SL-dependent shift of pCa50, i.e., ΔpCa50, was less pronounced (~0.11pCa units), with and without protein kinase A treatment. The rate of active force redevelopment was lower in ΔK210 preparations than in WT preparations, showing blunted thin filament cooperative activation. An increase in thin filament cooperative activation upon an increase in the fraction of strongly bound cross-bridges by MgADP increased ΔpCa50 to ~0.21pCa units. The depressed Frank-Starling mechanism in ΔK210 hearts is the result of a reduction in thin filament cooperative activation.


Asunto(s)
Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/fisiopatología , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/fisiopatología , Eliminación de Secuencia , Troponina T/genética , Adenosina Difosfato/metabolismo , Animales , Calcio/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Técnicas In Vitro , Ratones , Ratones Transgénicos , Contracción Miocárdica/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Troponina T/metabolismo
12.
Biochem Biophys Res Commun ; 440(4): 677-82, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24099767

RESUMEN

Glycogen synthase kinase (GSK)-3ß plays an important role in osteoblastogenesis by regulating the Wnt/ß-catenin signaling pathway. Therefore, we investigated whether GSK-3ß deficiency affects bone development and regeneration using mice heterozygously deficient for GSK-3ß (GSK-3ß(+/-)). The amounts of ß-catenin, c-Myc, cyclin D1, and runt-related transcription factor-2 (Runx2) in the bone marrow cells of GSK-3ß(+/-) mice were significantly increased compared with those of wild-type mice, indicating that Wnt/ß-catenin signals were enhanced in GSK-3ß(+/-) mice. Microcomputed tomography of the distal femoral metaphyses demonstrated that the volumes of both the cortical and trabecular bones were increased in GSK-3ß(+/-) mice compared with those in wild-type mice. Subsequently, to investigate the effect of GSK-3ß deficiency on bone regeneration, we established a partial bone defect in the femur and observed new bone at 14 days after surgery. The volume and mineral density of the new bone were significantly higher in GSK-3ß(+/-) mice than those in wild-type mice. These results suggest that bone formation and regeneration in vivo are accelerated by inhibition of GSK-3ß, probably through activation of the Wnt/ß-catenin signaling pathway.


Asunto(s)
Desarrollo Óseo , Regeneración Ósea , Glucógeno Sintasa Quinasa 3/metabolismo , Osteoblastos/fisiología , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Ciclina D1/metabolismo , Glucógeno Sintasa Quinasa 3/genética , Ratones , Osteoblastos/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal
13.
J Pharmacol Sci ; 121(2): 103-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23357875

RESUMEN

Differentiation-inducing factor-1 (DIF-1), a morphogen for Dictyostelium discoideum, inhibits the proliferation of human cancer cell lines by suppressing the Wnt/ß-catenin signaling pathway. In this study, we examined the effect of DIF-1 on c-Myc, a target gene product of the Wnt/ß-catenin signaling pathway, mainly using HCT-116 colon cancer cells. DIF-1 strongly reduced the amount of c-Myc protein in time- and concentration-dependent manners and reduced c-Myc mRNA expression by inhibiting promoter activity through the TCF binding sites. The effect of DIF-1 on c-Myc was also confirmed using the human cervical cell line HeLa. Pretreatment with the proteasome inhibitor MG132 or glycogen synthase kinase-3ß (GSK-3ß) inhibitors (LiCl and SB216763) attenuated the effect of DIF-1, suggesting that DIF-1 induced c-Myc protein degradation through GSK-3ß activation. Furthermore, we examined whether c-Myc was involved in the anti-proliferative effect of DIF-1 using c-Myc-overexpressing cells and found that c-Myc was associated with the anti-proliferative effect of this compound. These results suggest that DIF-1 inhibits c-Myc expression by inhibiting promoter activity and inducing protein degradation via GSK-3ß activation, resulting in the inhibition of cell proliferation. Since c-Myc seems to be profoundly involved in accelerated proliferation of various malignant tumors, DIF-1 may have a potential to develop into a novel anti-cancer agent.


Asunto(s)
Hexanonas/farmacología , Hidrocarburos Clorados/farmacología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Hexanonas/antagonistas & inhibidores , Humanos , Hidrocarburos Clorados/antagonistas & inhibidores , Indoles/farmacología , Leupeptinas/farmacología , Cloruro de Litio/farmacología , Maleimidas/farmacología , Inhibidores de Proteasoma/farmacología , Transducción de Señal/efectos de los fármacos
14.
J Mol Cell Cardiol ; 53(6): 760-7, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22921782

RESUMEN

Inherited or non-inherited dilated cardiomyopathy (DCM) patients develop varied disease phenotypes leading to death after developing congestive heart failure (HF) or sudden death with mild or no overt HF symptoms, suggesting that environmental and/or genetic factors may modify the disease phenotype of DCM. In this study, we sought to explore unknown genetic factors affecting the disease phenotype of monogenic inherited human DCM. Knock-in mice bearing a sarcomeric protein mutation that causes DCM were created on different genetic backgrounds; BALB/c and C57Bl/6. DCM mice on the BALB/c background showed cardiac enlargement and systolic dysfunction and developed congestive HF before died. In contrast, DCM mice on the C57Bl/6 background developed no overt HF symptoms and died suddenly, although they showed considerable cardiac enlargement and systolic dysfunction. BALB/c mice have brain serotonin dysfunction due to a single nucleotide polymorphism (SNP) in tryptophan hydroxylase 2 (TPH2). Brain serotonin dysfunction plays a critical role in depression and anxiety and BALB/c mice exhibit depression- and anxiety-related behaviors. Since depression is common and associated with poor prognosis in HF patients, we examined therapeutic effects of anti-depression drug paroxetine and anti-anxiety drug buspirone that could improve the brain serotonin function in mice. Both drugs reduced cardiac enlargement and improved systolic dysfunction and symptoms of severe congestive HF in DCM mice on the BALB/c background. These results strongly suggest that genetic backgrounds involving brain serotonin dysfunction, such as TPH2 gene SNP, may play an important role in the development of congestive HF in DCM.


Asunto(s)
Encéfalo/metabolismo , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Serotonina/metabolismo , Animales , Modelos Animales de Enfermedad , Corazón/efectos de los fármacos , Insuficiencia Cardíaca/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocardio/metabolismo , Miocardio/patología , Fenotipo , Agonistas del Receptor de Serotonina 5-HT1/farmacología
15.
Circ Heart Fail ; 15(12): e009366, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36268712

RESUMEN

BACKGROUND: Dilated cardiomyopathy (DCM) is a life-threatening disease, resulting in refractory heart failure. An immune disorder underlies the pathophysiology associated with heart failure progression. Invariant natural killer T (iNKT) cell activation is a prospective therapeutic strategy for ischemic heart disease. However, its efficacy in nonischemic cardiomyopathy, such as DCM, remains to be elucidated, and the feasible modality for iNKT cell activation in humans is yet to be validated. METHODS: Dendritic cells isolated from human volunteers were pulsed with α-galactosylceramide ex vivo, which were used as α-galactosylceramide-pulsed dendritic cells (αGCDCs). We treated DCM mice harboring mutated troponin TΔK210/ΔK210 with αGCDCs and evaluated the efficacy of iNKT cell activation on heart failure in DCM mice. Furthermore, we investigated the molecular basis underlying its therapeutic effects in these mice and analyzed primary cardiac cells under iNKT cell-secreted cytokines. RESULTS: The number of iNKT cells in the spleens of DCM mice was reduced compared with that in wild-type mice, whereas αGCDC treatment activated iNKT cells, prolonged survival of DCM mice, and prevented decline in the left ventricular ejection fraction for 4 weeks, accompanied by suppressed interstitial fibrosis. Mechanistically, αGCDC treatment suppressed TGF (transforming growth factor)-ß signaling and expression of fibrotic genes and restored vasculature that was impaired in DCM hearts by upregulating angiopoietin 1 (Angpt1) expression. Consistently, IFNγ (interferon gamma) suppressed TGF-ß-induced Smad2/3 signaling and the expression of fibrotic genes in cardiac fibroblasts and upregulated Angpt1 expression in cardiomyocytes via Stat1. CONCLUSIONS: Immunomodulatory cell therapy with αGCDCs is a novel therapeutic strategy for heart failure in DCM.


Asunto(s)
Cardiomiopatía Dilatada , Insuficiencia Cardíaca , Ratones , Humanos , Animales , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/terapia , Cardiomiopatía Dilatada/metabolismo , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/terapia , Insuficiencia Cardíaca/metabolismo , Volumen Sistólico , Función Ventricular Izquierda , Fibrosis , Células Dendríticas/metabolismo , Tratamiento Basado en Trasplante de Células y Tejidos
16.
Circulation ; 122(9): 891-9, 2010 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-20713897

RESUMEN

BACKGROUND: Dilated cardiomyopathy (DCM), characterized by dilatation and dysfunction of the left ventricle, is an important cause of heart failure. Many mutations in various genes, including cytoskeletal protein genes and contractile protein genes, have been identified in DCM patients, but the mechanisms of how such mutations lead to DCM remain unknown. METHODS AND RESULTS: We established the mouse model of DCM by expressing a mutated cardiac alpha-actin gene, which has been reported in patients with DCM, in the heart (mActin-Tg). mActin-Tg mice showed gradual dilatation and dysfunction of the left ventricle, resulting in death by heart failure. The number of apoptotic cardiomyocytes and protein levels of p53 were increased in the hearts of mActin-Tg mice. Overexpression of Bcl-2 or downregulation of p53 decreased the number of apoptotic cardiomyocytes and improved cardiac function. This mouse model showed a decrease in myofilament calcium sensitivity and activation of calcium/calmodulin-dependent kinase IIdelta (CaMKIIdelta). The inhibition of CaMKIIdelta prevented the increase in p53 and apoptotic cardiomyocytes and ameliorated cardiac function. CONCLUSIONS: CaMKIIdelta plays a critical role in the development of heart failure in part by accumulation of p53 and induction of cardiomyocyte apoptosis in the DCM mouse model.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Cardiomiopatía Dilatada/fisiopatología , Insuficiencia Cardíaca/fisiopatología , Proteína p53 Supresora de Tumor/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/genética , Animales , Apoptosis/fisiología , Bencilaminas/farmacología , Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Modelos Animales de Enfermedad , Activación Enzimática/fisiología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Humanos , Ratones , Ratones Transgénicos , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología , Inhibidores de Proteínas Quinasas/farmacología , Sulfonamidas/farmacología
17.
Biochem Biophys Res Commun ; 409(1): 108-13, 2011 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-21565173

RESUMEN

Dilated cardiomyopathy (DCM) is a myocardial disorder that is characterized by dilation and dysfunction of the left ventricle (LV). Accumulating evidence has implicated aberrant Ca(2+) signaling and oxidative stress in the progression of DCM, but the molecular details are unknown. In the present study, we report that inhibition of the transient receptor potential canonical 3 (TRPC3) channels partially prevents LV dilation and dysfunction in muscle LIM protein-deficient (MLP (-/-)) mice, a murine model of DCM. The expression level of TRPC3 and the activity of Ca(2+)/calmodulin-dependent kinase II (CaMKII) were increased in MLP (-/-) mouse hearts. Acitivity of Rac1, a small GTP-binding protein that participates in NADPH oxidase (Nox) activation, and the production of reactive oxygen species (ROS) were also increased in MLP (-/-) mouse hearts. Treatment with pyrazole-3, a TRPC3 selective inhibitor, strongly suppressed the increased activities of CaMKII and Rac1, as well as ROS production. In contrast, activation of TRPC3 by 1-oleoyl-2-acetyl-sn-glycerol (OAG), or by mechanical stretch, induced ROS production in rat neonatal cardiomyocytes. These results suggest that up-regulation of TRPC3 is responsible for the increase in CaMKII activity and the Nox-mediated ROS production in MLP (-/-) mouse cardiomyocytes, and that inhibition of TRPC3 is an effective therapeutic strategy to prevent the progression of DCM.


Asunto(s)
Calcio/metabolismo , Cardiomiopatía Dilatada/metabolismo , Miocardio/metabolismo , Neuropéptidos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Canales Catiónicos TRPC/metabolismo , Disfunción Ventricular Izquierda/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Cardiomiopatía Dilatada/genética , Proteínas con Dominio LIM , Ratones , Ratones Mutantes , Proteínas Musculares/genética , Pirazoles/farmacología , Ratas , Canales Catiónicos TRPC/antagonistas & inhibidores , Disfunción Ventricular Izquierda/genética , Proteína de Unión al GTP rac1
18.
J Pharmacol Exp Ther ; 338(1): 2-11, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21430081

RESUMEN

Celecoxib, a cyclooxygenase-2 (COX-2)-selective nonsteroidal anti-inflammatory drug, has been shown to inhibit Akt and prevent cardiac remodeling in aortic banding-induced failing heart in mice. However, it may be difficult to use celecoxib for the treatment of heart failure because of thromboembolic adverse reactions. Since 2,5-dimethyl (DM)-celecoxib, a derivative unable to inhibit COX-2, has been also reported to inhibit Akt, we attempted to examine whether DM-celecoxib retains the ability to prevent cardiac remodeling and improve cardiac functions using a mouse model of inherited dilated cardiomyopathy (DCM). DM-celecoxib as well as celecoxib administered daily for 4 weeks inhibited Akt and subsequent phosphorylation of glycogen synthase kinase-3ß and mammalian target of rapamycin. Furthermore, both celecoxib and DM-celecoxib inhibited the activities of nuclear factor of activated T cell and ß-catenin and the expression of TCF7L2 (T-cell-specific transcriptional factor-7L2) and c-Myc, downstream mediators related to cardiac hypertrophy. Functional and morphological measurements showed that these compounds improved left ventricular systolic functions (ejection fraction: vehicle, 34.7 ± 3.9%; 100 mg/kg celecoxib, 50.3 ± 1.1%, p < 0.01; 100 mg/kg DM-celecoxib, 49.8 ± 0.8%, p < 0.01), which was also evidenced by the decrease in ß-myosin heavy chain and B-type natriuretic peptide, and prevented hypertrophic cardiac remodeling (heart/body weight ratio: vehicle, 10.4 ± 0.7 mg/g; 100 mg/kg celecoxib, 8.0 ± 0.3 mg/g, p < 0.01; 100 mg/kg DM-celecoxib, 8.2 ± 0.1 mg/g, p < 0.05). As a consequence, both compounds improved the survival rate (vehicle, 45%; 100 mg/kg celecoxib, 75%, p < 0.05; 100 mg/kg DM-celecoxib, 70%, p < 0.05). These results suggested that not only celecoxib but also DM-celecoxib prevents cardiac remodeling and reduces mortality in DCM through a COX-2-independent mechanism involving Akt and its downstream mediators.


Asunto(s)
Cardiomiopatía Dilatada/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Pirazoles/farmacología , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología , Remodelación Ventricular/efectos de los fármacos , Animales , Cardiomiopatía Dilatada/enzimología , Cardiomiopatía Dilatada/genética , Celecoxib , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Técnicas de Sustitución del Gen , Ratones , Proteínas Proto-Oncogénicas c-akt/fisiología , Pirazoles/uso terapéutico , Transducción de Señal/genética , Sulfonamidas/uso terapéutico , Remodelación Ventricular/genética
19.
J Pharmacol Sci ; 115(4): 490-9, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21422729

RESUMEN

Polymorphisms at codons 49 and 389 of the ß(1)-adrenergic receptor gene have been shown to alter the receptor function in vitro, whereas it remains controversial whether they influence the response to ß-blocker in vivo. In the present study, we investigated whether these polymorphisms influence the acute changes of heart rate and blood pressure induced by the ß(1)-adrenergic receptor-selective blocker atenolol in healthy young Japanese. A double-blind study was conducted with 307 subjects randomly allocated 2:1 to atenolol (50 mg) or placebo groups. Heart rate and blood pressure were significantly reduced after administration of atenolol in comparison to the placebo. In 207 subjects allocated to the atenolol group, the numbers of Ser/Ser, Ser/Gly, and Gly/Gly allele carriers for codon 49 were 159, 46, and 2, respectively; and those of Arg/Arg, Arg/Gly, and Gly/Gly for codon 389 were 129, 66, and 12, respectively. No significant association was identified between the changes in heart rate or blood pressure and either of the two polymorphisms. There was also no difference in the changes in heart rate or blood pressure among the diplotypes. The results of the present study do not support clinical use of genotyping for these polymorphisms to predict responses to ß-blockers.


Asunto(s)
Antagonistas de Receptores Adrenérgicos beta 1/farmacología , Pueblo Asiatico/genética , Atenolol/farmacología , Polimorfismo de Longitud del Fragmento de Restricción , Receptores Adrenérgicos beta 1/genética , Adulto , Alelos , Atenolol/sangre , Presión Sanguínea/efectos de los fármacos , Femenino , Genotipo , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Masculino
20.
Sci Rep ; 11(1): 14698, 2021 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-34282197

RESUMEN

In contrast to hypertrophic cardiomyopathy, there has been reported no specific pattern of cardiomyocyte array in dilated cardiomyopathy (DCM), partially because lack of alignment assessment in a three-dimensional (3D) manner. Here we have established a novel method to evaluate cardiomyocyte alignment in 3D using intravital heart imaging and demonstrated homogeneous alignment in DCM mice. Whilst cardiomyocytes of control mice changed their alignment by every layer in 3D and position twistedly even in a single layer, termed myocyte twist, cardiomyocytes of DCM mice aligned homogeneously both in two-dimensional (2D) and in 3D and lost myocyte twist. Manipulation of cultured cardiomyocyte toward homogeneously aligned increased their contractility, suggesting that homogeneous alignment in DCM mice is due to a sort of alignment remodelling as a way to compensate cardiac dysfunction. Our findings provide the first intravital evidence of cardiomyocyte alignment and will bring new insights into understanding the mechanism of heart failure.


Asunto(s)
Cardiomiopatía Dilatada/diagnóstico por imagen , Movimiento Celular/fisiología , Miocitos Cardíacos/fisiología , Animales , Animales Recién Nacidos , Cardiomiopatía Dilatada/patología , Cardiomiopatía Hipertrófica/diagnóstico por imagen , Cardiomiopatía Hipertrófica/patología , Células Cultivadas , Diagnóstico por Imagen/métodos , Masculino , Ratones , Ratones Transgénicos , Miocitos Cardíacos/citología , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA