Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Herz ; 40(5): 783-7, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25900768

RESUMEN

BACKGROUND: Mitochondrial coupling factor 6 (CF6) is a constriction factor in cardiac hypertrophy, whose mechanisms are not fully understood. MATERIALS AND METHODS: Here, we established cardiac hypertrophy models for feeding spontaneously hypertensive rats (SHRs) aged 10, 20, and 30 weeks. Hemodynamic monitoring was performed during the feeding program to ensure the success of the model. RESULTS: Cardiac hypertrophy, but not fibrosis, occurred in the 10-, 20-, and 30-week-old SHRs. No significant changes in CF6 gene expression were detected by RT-PCR in any of the SHR groups as compared with the control groups (p > 0.05). ELISA assessment showed that the CF6 protein level in the 20- and 30-week-old SHRs with cardiac hypertrophy was significantly increased (vs. control, p < 0.05). CONCLUSION: CF6 protein was upregulated in cardiac hypertrophy induced by hypertension; further mechanisms involved in this process should be investigated.


Asunto(s)
Cardiomegalia/etiología , Cardiomegalia/metabolismo , Hipertensión/complicaciones , Hipertensión/metabolismo , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Factores de Acoplamiento de la Fosforilación Oxidativa/metabolismo , Animales , Biomarcadores/metabolismo , Masculino , Ratas Endogámicas SHR , Ratas Wistar
2.
Int J Mol Sci ; 14(3): 4805-16, 2013 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-23449031

RESUMEN

Granulocyte-colony stimulating factor (G-CSF) has been reported to improve the function of infarcted heart, but its effects on atherosclerosis are unclear. Here we examined the effects and the potential mechanisms in the high-fat diet rabbit model. Six-month-old male New Zealand white rabbits, fed a high-cholesterol diet or a normal diet for 10 weeks, were treated with vehicle or G-CSF. G-CSF increased lesion area in the thoracic aorta and the plasma levels of total cholesterol (TC) and low-density lipoprotein-cholesterol (LDL-C) at the early phase in the high-fat diet group. High-fat diet-induced arterial endothelium damage and apoptosis were greatly aggravated by G-CSF treatment. In vivo, G-CSF impaired apoptosis induced by oxidized low density lipoprotein (OX-LDL) but it had little effect on cultured endothelial cells (ECs) with vehicle treatment. Further research revealed that G-CSF promoted the upregulation of endothelin-1 (ET-1) and the downregulation of endothelial nitric oxide synthase (eNOS) of thoracic aortae induced by a high-fat diet. In vitro, the effects of G-CSF on expression of ET-1 and eNOS in cultured ECs were consistent with those in vivo. Our results suggested that G-CSF exacerbates lipid abnormity and endothelium damage in hyperlipidemia rabbits, thereby resulting in the deterioration of atherosclerosis and that the ET-1/eNOS system may regulate the progression.

3.
J Biomed Biotechnol ; 2012: 737134, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22500105

RESUMEN

AngiotensinII (AngII) is involved in not only the formation of cardiac hypertrophy but also the development of cardiac remodeling both of which are associated with myocardial angiogenesis. This study was therefore performed to clarify the effects of AngII on the formation of vasculatures by cultured cardiac microvascular endothelial cells (CMVECs) after a long-period stimulation with or without the AngII preconditioning. Incubation with AngII for 18 hrs significantly impaired the formation of capillary-like tubes comparing to that without AngII. CMVECs with AngII pretreatment for 5 and 10 min formed more capillary-like tubes than those without AngII pretreatment, suggesting that preconditioning with AngII at a lower dose for a short period could prevent the further damage of CMVECs by a higher concentration of AngII. Moreover, AngII (10(-7) M) stimulation for 5 and 10 min significantly induced the increase in extracellular signal-regulated protein kinases (ERKs) phosphorylation, and an ERKs inhibitor, PD98059, abrogated the increase in the formation of capillary-like tubes induced by the AngII-pretreatment. In conclusion, preconditioning with a lower concentration of AngII for a short period prevents the subsequent impairment of CMVECs by a higher dose of AngII, at least in part, through the increase in ERKs phosphorylation.


Asunto(s)
Angiotensina II/farmacología , Células Endoteliales/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Análisis de Varianza , Inductores de la Angiogénesis/farmacología , Animales , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Flavonoides/farmacología , Masculino , Miocardio/citología , Fosforilación , Sustancias Protectoras/farmacología , Ratas , Ratas Wistar
4.
J Ultrasound Med ; 31(7): 1111-21, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22733860

RESUMEN

OBJECTIVES: Animal models of myocardial infarction (MI) are widely used not only in analyses of the mechanisms but also in testing the efficacy of therapeutic strategies for the disease. It is therefore critically important but almost impossible to exactly evaluate the validity of coronary artery ligation in a mouse model of MI except by anatomic and histologic analyses. We explored a noninvasive method to estimate MI through analyses of coronary perfusion by transthoracic echocardiography in mice before and 1 day after ligation of the left anterior descending coronary artery. METHODS: Transthoracic echocardiography-based cardiac function, geometry, and coronary perfusion, electrocardiographic findings, and serum troponin I levels were examined in C57BL6/J mice subjected to left anterior descending artery ligation. The histologic infarct size was confirmed by staining the heart with 2,3,5-triphenyltetrazolium chloride. RESULTS: Among all parameters, the postoperative hyperemic peak diastolic velocity and coronary flow reserve were most correlated with infarct size (R² = .8028 and .5825, respectively; both P < .0001). With an infarct size of 30% or greater indicating successful ligation and less than 30% indicating unsuccessful ligation, receiver operating characteristic curve analysis showed that the postoperative hyperemic peak diastolic velocity and coronary flow reserve most effectively indicated the infarct size level with optimal cutoff values of 480.16 mm/s and 1.89, respectively. Furthermore, impaired cardiac function, an eccentrically expanded left ventricle, typical pathologic electrocardiographic findings, and elevated troponin I levels were observed most often in the mice with an impaired hyperemic peak diastolic velocity and coronary flow reserve. CONCLUSIONS: The echocardiographic hyperemic peak diastolic velocity and coronary flow reserve can estimate the histologic infarct size in mice with coronary occlusion.


Asunto(s)
Estenosis Coronaria/diagnóstico por imagen , Modelos Animales de Enfermedad , Ecocardiografía/métodos , Infarto del Miocardio/diagnóstico por imagen , Imagen de Perfusión Miocárdica/métodos , Animales , Estenosis Coronaria/complicaciones , Humanos , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/etiología , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
5.
J Mol Cell Cardiol ; 51(5): 821-9, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21854784

RESUMEN

Heat shock transcription factor 1 (HSF1) plays an important role not only in excise-induced cardiac hypertrophy but also in protection against pressure overload-induced cardiac dysfunction. However, the mechanism is not completely understood. We here elucidate the potential mechanisms by which HSF1 protects against pressure overload-induced cardiac remodeling and dysfunction. A sustained constriction of transverse aorta (TAC) was imposed to HSF1 transgenic (TG), knockout (KO) and their littermate wild type (WT) male mice. Four weeks later, adaptive responses to TAC, such as cardiac hypertrophy, contractility and angiogenesis evaluated by echocardiography, catheterization, coronary perfusion pressure and immunohistochemistry were well preserved in TG but not in KO compared with WT mice. An angiogenesis inhibitor TNP-470 abrogated all these adaptive responses in TG mice, while cardiac transfection of VEGF with angiopoietin-1 rescued the broken heart in KO mice. In response to TAC, p53 was downregulated and hypoxia-inducing transcription factor-1 (HIF-1) was upregulated not only in the heart but also in the cultured cardiac endothelial cells (EC) of TG mice as compared to WT mice whereas these changes became opposite in KO mice. A small interfering RNA (siRNA) of HIF-1 but not a p53 gene impaired the adaptive responses of the heart and EC in TG mice, and a siRNA of p53 but not a HIF-1 gene significantly reversed the heart and EC disorders in KO mice after TAC. We conclude that HSF1 promotes cardiac angiogenesis through suppression of p53 and subsequent upregulation of HIF-1 in endothelial cells during chronic pressure overload, leading to the maintenance of cardiac adaptation.


Asunto(s)
Angiopoyetina 1/metabolismo , Cardiomegalia/metabolismo , Proteínas de Unión al ADN/metabolismo , Células Endoteliales/metabolismo , Miocitos Cardíacos/metabolismo , Neovascularización Fisiológica/genética , Factores de Transcripción/metabolismo , Disfunción Ventricular Izquierda/metabolismo , Inhibidores de la Angiogénesis/efectos adversos , Angiopoyetina 1/genética , Animales , Cardiomegalia/diagnóstico por imagen , Cardiomegalia/etiología , Cardiomegalia/genética , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Células Cultivadas , Constricción , Ciclohexanos/efectos adversos , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Ecocardiografía , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Factores de Transcripción del Choque Térmico , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , O-(Cloroacetilcarbamoil) Fumagilol , Presión/efectos adversos , ARN Interferente Pequeño/farmacología , Sesquiterpenos/efectos adversos , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Transfección , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Disfunción Ventricular Izquierda/diagnóstico por imagen , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/fisiopatología
6.
Int J Mol Sci ; 12(2): 1306-15, 2011 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-21541060

RESUMEN

This paper explores the effect of granulocyte colony-stimulating factor (G-CSF) on mouse myocardial microvascular endothelial cell (CMECs) proliferation. CMECs were harvested from C57/BL6 mice. CMECs were cultured in medium containing G-CSF (0 ng/mL, 20 ng/mL, 40 ng/mL, 60 ng/mL) for five days. Proliferative activity of CMECs was examined by CCK-8 method. Hypoxia inducible factor-1 (HIF-1) and p53 expression levels was determined from the mRNA obtained by reverse transcription polymerase chain reaction (RT-PCR). Results showed that the purity quotient of the CMECs, which were cultured by the method of modified myocardial tissue explant culture, was higher than 95%. Compared with control untreated cells, the proliferative activity of CMECs and the expression level of HIF-1 mRNA in these cells were enhanced by G-CSF treatment, whereas the expression level of p53 mRNA was markedly reduced. It may be concluded that G-CSF could promote the proliferative activity of CMECs, which might be mediated by upregulation of HIF-1 and downregulation of p53.


Asunto(s)
Proliferación Celular , Células Endoteliales/metabolismo , Factor Estimulante de Colonias de Granulocitos/efectos de los fármacos , Animales , Células Cultivadas , Vasos Coronarios/citología , Regulación hacia Abajo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Factor 1 Inducible por Hipoxia/genética , Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Endogámicos C57BL , Microvasos/citología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
7.
J Colloid Interface Sci ; 586: 836-846, 2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33208244

RESUMEN

Manganese-cerium metal oxide with flocculent structure prepared via the pyrolysis of Mn/Ce-MOF and supported Pd were applied for the catalytic oxidation of toluene. The Pd/Mn3Ce2-300 catalyst could completely oxidize toluene at 190 °C, which presented excellent catalytic performance. Moreover, Pd/Mn3Ce2-300 possessed great reusability, stability and water resistance even under 10 vol% water vapors. A series of characterizations including X-ray diffraction (XRD), N2 adsorption-desorption, scanning electron microscopy (SEM), X-ray photoelectron spectroscopy (XPS) and H2 temperature programmed reduction (H2-TPR) were used to investigate the physicochemical properties of the samples. It was found that Pd/Mn3Ce2-300 possessed a better reduction ability at low temperature, more surface absorbed oxygen and surface Pd species, and a strong interaction between Pd and Mn3Ce2-300, resulting in great catalytic performance for toluene degradation.

8.
Curr Neurovasc Res ; 14(1): 11-18, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28382863

RESUMEN

Peroxisome proliferator-activated receptors alpha (PPARα) is a therapy target in atherosclerosis and cardiovascular diseases. However, anti-inflammatory effects of PPARα in intracerebral hemorrhage (ICH) remain unknown. We investigated the anti-inflammatory effects of fenofibrate, a ligand of PPARα, in ICH rat model. We found that engagement of fenofibrate increased nissl body and astrocytes, and reduced the neuronal damage, which was observed in paraffin section of ICH rat brain. Fenofibrate also promoted the proliferation of astrocytes that were isolated from adult rat brain. Fenofibrate significantly upregulated heme oxygenase 1 (HO-1) at protein and mRNA levels in human glioblastoma LN-18 cells and rat brain astrocytes respectively, but nuclear factor kappalight- chain-enhancer of activated B cells (NFκB) was downregulated after fenofibrate treatment. Results showed that fenofibrate-induced upregulation of HO-1 expression were inhibited after LN-18 cells were transfected with 50nM small interfering RNA (siRNAs) for 48 hours to knockdown PPARα. Further studies in rat astrocytes confirmed the rescue effects of PPARα silence against fenofibrate induced upregulation of HO-1 expression. Our data indicated that fenofibrate benefits neuronal protection through increasing HO-1 expression level and decreasing NFκB expression in PPARα-dependent manner. In conclusion, PPARα and HO-1 may function as significant targets to protect the brain during ICH.


Asunto(s)
Astrocitos/efectos de los fármacos , Encéfalo/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Hemorragia Cerebral/tratamiento farmacológico , Fenofibrato/farmacología , Hemo-Oxigenasa 1/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Encéfalo/metabolismo , Células Cultivadas , Hemo-Oxigenasa 1/genética , Masculino , PPAR alfa/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Ratas Sprague-Dawley
9.
Chin Med J (Engl) ; 130(3): 328-333, 2017 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-28139517

RESUMEN

BACKGROUND: Angiotensin II (Ang II) is a major contributor to the development of heart failure. However, the molecular and cellular mechanisms that underlie this process remain elusive. Inadequate angiogenesis in the myocardium leads to a transition from cardiac hypertrophy to dysfunction, and our previous study showed that Ang II significantly impaired the angiogenesis response. The current study was designed to examine the role of Jagged1-Notch signaling in the effect of Ang II during impaired angiogenesis and cardiac hypertrophy. METHODS: Ang II was subcutaneously infused into 8-week-old male C57BL/6 mice at a dose of 200 ng·kg-1·min-1 for 2 weeks using Alzet micro-osmotic pumps. N-[N-(3, 5-difluorophenacetyl)-L-alanyl]-S-phenylglycine tert-butyl ester (DAPT), a γ-secretase inhibitor, was injected subcutaneously during Ang II infusion at a dose of 10.0 mg·kg-1·d-1. Forty mice were divided into four groups (n = 10 per group): control group; Ang II group, treated with Ang II; DAPT group, treated with DAPT; and Ang II + DAPT group, treated with both Ang II and DAPT. At the end of experiments, myocardial (left ventricle [LV]) tissue from each experimental group was evaluated using immunohistochemistry, Western blotting, and real-time polymerase chain reaction. Data were analyzed using one-way analysis of variance test followed by the least significant difference method or independent samples t-test. RESULTS: Ang II treatment significantly induced cardiac hypertrophy and impaired the angiogenesis response compared to controls, as shown by hematoxylin and eosin (HE) staining and immunohistochemistry for CD31, a vascular marker (P < 0.05 for both). Meanwhile, Jagged1 protein was significantly increased, but gene expression for both Jag1 and Hey1 was decreased in the LV following Ang II treatment, compared to that in controls (relative ratio for Jag1 gene: 0.45 ± 0.13 vs. 0.84 ± 0.15; relative ratio for Hey1 gene: 0.51 ± 0.08 vs. 0.91 ± 0.09; P < 0.05). All these cellular and molecular effects induced by Ang II in the hearts of mice were reduced by DAPT treatment. Interestingly, Ang II stimulated Hey1, a known Notch target, but did not affect the expression of Hey2, another Notch target gene. CONCLUSIONS: A Jagged1-Hey1 signal might mediate the impairment of angiogenesis induced by Ang II during cardiac hypertrophy.


Asunto(s)
Cardiomegalia/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteína Jagged-1/metabolismo , Miocardio/metabolismo , Animales , Cardiomegalia/inducido químicamente , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
10.
PLoS One ; 8(10): e76529, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24098521

RESUMEN

Angiotensin II (AngII) is a major contributor to the development of heart failure, however, the molecular and cellular mechanisms still remain elucidative. Inadequate angiogenesis in myocardium leads to transition from cardiac hypertrophy to dysfunction, this study was therefore conducted to examine the effects of AngII on myocardial angiogenesis and the underlying mechanisms. AngII treatment significantly impaired angiogenetic responses, which were determined by counting the capillaries either in matrigel formed by cultured cardiac microvascular endothelial cells (CMVECs) or in myocardium of mice and by measuring the in vitro and in vivo production of VEGF proteins, and stimulated accumulation and phosphorylation of cytosolic p53 which led to increases in phosphorylated p53 and decreases of hypoxia inducible factor (Hif-1) in nucleus. All of these cellular and molecular events induced by AngII in CEMCs and hearts of mice were largely reduced by a p53 inhibitor, pifithrin-α (PFT-α). Interestingly, AngII stimulated the upregulation of Jagged1, a ligand of Notch, but it didn't affect the expression of Delta-like 4 (Dll-4), another ligand of Notch. Inhibition of p53 by PFT-α partly abolished this effect of AngII. Further experiments showed that knockdown ofJagged1 by addition of siRNA to cultured CMVECs dramatically declined AngII-stimulated accumulation and phosphorylation of p53 in cytosol, upregulation of phosphorylated p53 and downregulation of Hif-1 expression in nucleus, decrease of VEGF production and impairment of capillary-like tube formation by the cells. Our data collectively suggest that AngII impairs myocardial angiogenetic responses through p53-dependent downregulation of Hif-1 which is regulated by Jagged1/Notch1 signaling.


Asunto(s)
Angiotensina II/farmacología , Proteínas de Unión al Calcio/genética , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de la Membrana/genética , Neovascularización Fisiológica/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética , Animales , Benzotiazoles/farmacología , Proteínas de Unión al Calcio/metabolismo , Núcleo Celular/metabolismo , Colágeno/química , Combinación de Medicamentos , Células Endoteliales/citología , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Regulación de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Bombas de Infusión Implantables , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Jagged-1 , Laminina/química , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Miocardio/citología , Miocardio/metabolismo , Cultivo Primario de Células , Proteoglicanos/química , Ratas , Ratas Wistar , Receptor Notch1/genética , Receptor Notch1/metabolismo , Proteínas Serrate-Jagged , Transducción de Señal , Tolueno/análogos & derivados , Tolueno/farmacología , Proteína p53 Supresora de Tumor/metabolismo
11.
Hypertension ; 58(6): 1099-110, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21986507

RESUMEN

Ryanodine receptor type 2 (RyR-2) mediates Ca(2+) release from sarcoplasmic reticulum and contributes to myocardial contractile function. However, the role of RyR-2 in the development of cardiac hypertrophy is not completely understood. Here, mice with or without reduction of RyR-2 gene (RyR-2(+/-) and wild-type, respectively) were analyzed. At baseline, there was no difference in morphology of cardiomyocyte and heart and cardiac contractility between RyR-2(+/-) and wild-type mice, although Ca(2+) release from sarcoplasmic reticulum was impaired in isolated RyR-2(+/-) cardiomyocytes. During a 3-week period of pressure overload, which was induced by constriction of transverse aorta, isolated RyR-2(+/-) cardiomyocytes displayed more reduction of Ca(2+) transient amplitude, rate of an increase in intracellular Ca(2+) concentration during systole, and percentile of fractional shortening, and hearts of RyR-2(+/-) mice displayed less compensated hypertrophy, fibrosis, and contractility; more apoptosis with less autophagy of cardiomyocytes; and similar decrease of angiogenesis as compared with wild-type ones. Moreover, constriction of transverse aorta-induced increases in the activation of calcineurin, extracellular signal-regulated protein kinases, and protein kinase B/Akt but not that of Ca(2+)/calmodulin-dependent protein kinase II, and its downstream targets in the heart of wild-type mice were abolished in the RyR-2(+/-) one, suggesting that RyR-2 is a regulator of calcineurin, extracellular signal-regulated protein kinases, and Akt but not of calmodulin-dependent protein kinase II activation during pressure overload. Taken together, our data indicate that RyR-2 contributes to the development of cardiac hypertrophy and adaptation of cardiac function during pressure overload through regulation of the sarcoplasmic reticulum Ca(2+) release; activation of calcineurin, extracellular signal-regulated protein kinases, and Akt; and cardiomyocyte survival.


Asunto(s)
Hipertrofia Ventricular Izquierda/fisiopatología , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Animales , Apoptosis , Autofagia , Calcineurina/metabolismo , Señalización del Calcio , Tamaño de la Célula , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Perfilación de la Expresión Génica , Hemodinámica , Hipertrofia Ventricular Izquierda/diagnóstico por imagen , Hipertrofia Ventricular Izquierda/etiología , Hipertrofia Ventricular Izquierda/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Musculares/biosíntesis , Proteínas Musculares/genética , Contracción Miocárdica/fisiología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Presión/efectos adversos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/deficiencia , Canal Liberador de Calcio Receptor de Rianodina/genética , Retículo Sarcoplasmático/metabolismo , Ultrasonografía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA