Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
1.
Cardiovasc Res ; 118(2): 517-530, 2022 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-33705529

RESUMEN

AIMS: Hypertrophic cardiomyopathy (HCM) is characterized by cardiomyocyte hypertrophy and disarray, and myocardial stiffness due to interstitial fibrosis, which result in impaired left ventricular filling and diastolic dysfunction. The latter manifests as exercise intolerance, angina, and dyspnoea. There is currently no specific treatment for improving diastolic function in HCM. Here, we investigated whether myeloperoxidase (MPO) is expressed in cardiomyocytes and provides a novel therapeutic target for alleviating diastolic dysfunction in HCM. METHODS AND RESULTS: Human cardiomyocytes derived from control-induced pluripotent stem cells (iPSC-CMs) were shown to express MPO, with MPO levels being increased in iPSC-CMs generated from two HCM patients harbouring sarcomeric mutations in the MYBPC3 and MYH7 genes. The presence of cardiomyocyte MPO was associated with higher chlorination and peroxidation activity, increased levels of 3-chlorotyrosine-modified cardiac myosin binding protein-C (MYBPC3), attenuated phosphorylation of MYBPC3 at Ser-282, perturbed calcium signalling, and impaired cardiomyocyte relaxation. Interestingly, treatment with the MPO inhibitor, AZD5904, reduced 3-chlorotyrosine-modified MYBPC3 levels, restored MYBPC3 phosphorylation, and alleviated the calcium signalling and relaxation defects. Finally, we found that MPO protein was expressed in healthy adult murine and human cardiomyocytes, and MPO levels were increased in diseased hearts with left ventricular hypertrophy. CONCLUSION: This study demonstrates that MPO inhibition alleviates the relaxation defect in hypertrophic iPSC-CMs through MYBPC3 phosphorylation. These findings highlight cardiomyocyte MPO as a novel therapeutic target for improving myocardial relaxation associated with HCM, a treatment strategy which can be readily investigated in the clinical setting, given that MPO inhibitors are already available for clinical testing.


Asunto(s)
Cardiomiopatía Hipertrófica/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Hipertrofia Ventricular Izquierda/tratamiento farmacológico , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Peroxidasa/antagonistas & inhibidores , Función Ventricular Izquierda/efectos de los fármacos , Animales , Miosinas Cardíacas/genética , Miosinas Cardíacas/metabolismo , Cardiomiopatía Hipertrófica/enzimología , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/fisiopatología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Humanos , Hipertrofia Ventricular Izquierda/enzimología , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/fisiopatología , Células Madre Pluripotentes Inducidas/enzimología , Células Madre Pluripotentes Inducidas/patología , Masculino , Ratones Endogámicos C57BL , Mutación Missense , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Peroxidasa/metabolismo , Fosforilación , Especies Reactivas de Oxígeno/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo
2.
Biosci Rep ; 41(12)2021 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-34750628

RESUMEN

BACKGROUND: Angiotensin-converting enzyme (ACE) gene polymorphisms have recently been shown to be associated with risk of developing left ventricular hypertrophy (LVH). However, the results were controversial. We aimed to conduct this meta-analysis to further confirm the association between ACE rs4646994 polymorphism and hypertrophic cardiomyopathy (HCM)/dilated cardiomyopathy (DCM). METHODS: PubMed, Embase, the Chinese National Knowledge Information, and Wanfang databases were searched for eligible studies. The Newcastle-Ottawa Scale (NOS) was used to evaluate the quality of included studies. Then we evaluated the association between ACE gene mutation and HCM/DCM by calculating odds ratios (ORs) and 95% confidence intervals (95% CIs). Subgroup analysis was further performed to explore situations in specialized subjects. Sensitivity analysis and publication bias was assessed to confirm the study reliability. RESULTS: There were 13 studies on DCM (2004 cases and 1376 controls) and 16 studies on HCM (2161 controls and 1192 patients). ACE rs4646994 polymorphism was significantly associated with DCM in all genetic models. However, in HCM, four genetic models (allele model, homozygous model, heterozygous model, and dominant model) showed significant association between ACE rs4646994 polymorphism and DCM. In subgroup analysis, we found that ACE rs4646994 polymorphism was significantly associated with DCM/HCM in Asian population. Finally, we also conducted a cumulative meta-analysis, which indicates that the results of our meta-analysis are highly reliable. CONCLUSION: ACE rs4646994 polymorphism increases the risk of DCM/HCM in Asians, but not in Caucasians. More case-control studies are needed to strengthen our conclusions and to assess the gene-gene and gene-environment interactions between ACE rs4646994 polymorphism and DCM/HCM.


Asunto(s)
Cardiomiopatía Dilatada/genética , Cardiomiopatía Hipertrófica/genética , Peptidil-Dipeptidasa A/genética , Polimorfismo de Nucleótido Simple , Pueblo Asiatico/genética , Cardiomiopatía Dilatada/enzimología , Cardiomiopatía Dilatada/etnología , Cardiomiopatía Hipertrófica/enzimología , Cardiomiopatía Hipertrófica/etnología , Estudios de Casos y Controles , Interacción Gen-Ambiente , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Medición de Riesgo , Factores de Riesgo , Población Blanca/genética
3.
Ann Thorac Surg ; 111(3): 915-922, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32738221

RESUMEN

BACKGROUND: Myocardial fibrosis (MF) is considered a result of microvascular dysfunction in patients with hypertrophic cardiomyopathy. Matrix metalloproteinases (MMPs) and their tissue inhibitors (TIMPs), capable of degrading collagen, directly participate in the development of MF. First we investigated the relationships among MF, microvascular rarefaction, and MMPs. Then we assessed the prognostic value of MF-related circulating biomarkers. METHODS: Fifty-five obstructive hypertrophic cardiomyopathy (HOCM) patients were enrolled after surgical myectomy. Myocardial samples were performed with Masson's trichrome staining and immunohistochemical procedures for collagen volume fraction and microvascular density, respectively. Enzyme-linked immunosorbent assays were used to assess myocardial and plasma of MMP-2, MMP-9, and TIMP-1 and plasma C-terminal propeptide of procollagen type Ⅰ (PICP) and C-terminal telopeptide of type Ⅰ collagen (ICTP) levels. The composite cardiovascular endpoint consisted of new-onset atrial fibrillation, heart failure requiring hospitalization, and all-cause death. RESULTS: In HOCM patients microvascular density was associated with the myocardial MMP-2/TIMP-1 ratio (r = -0.348, P = .009), whereas no correlation was found between collagen volume fraction and myocardial MMPs. During the 44-month follow-up 6 patients experienced a cardiovascular endpoint. The plasma PICP/ICTP ratio and MMP-2/TIMP-1 ratio were the 2 strongest prognostic makers. In multivariable analyses high PICP/ICTP and MMP-2/TIMP-1 ratios remained independent predictors of cardiovascular outcomes after adjusting for clinical confounders (hazard ratios, 12.683 [P = .021] and 17.037 [P = .027], respectively). CONCLUSIONS: In HOCM patients the myocardial MMP-2/TIMP-1 ratio was elevated because of microvascular rarefaction but may not be responsible for MF. High plasma PICP/ICTP and MMP-2/TIMP-1 ratios are independent predictors of adverse outcomes in HOCM patients.


Asunto(s)
Cardiomiopatía Hipertrófica/enzimología , Circulación Coronaria/fisiología , Metaloproteinasas de la Matriz/sangre , Biomarcadores/sangre , Cardiomiopatía Hipertrófica/diagnóstico , Cardiomiopatía Hipertrófica/fisiopatología , Femenino , Estudios de Seguimiento , Humanos , Imagen por Resonancia Cinemagnética , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos
4.
Am J Physiol Heart Circ Physiol ; 320(2): H881-H890, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33337957

RESUMEN

Morbidity and mortality associated with heart disease is a growing threat to the global population, and novel therapies are needed. Mavacamten (formerly called MYK-461) is a small molecule that binds to cardiac myosin and inhibits myosin ATPase. Mavacamten is currently in clinical trials for the treatment of obstructive hypertrophic cardiomyopathy (HCM), and it may provide benefits for treating other forms of heart disease. We investigated the effect of mavacamten on cardiac muscle contraction in two transgenic mouse lines expressing the human isoform of cardiac myosin regulatory light chain (RLC) in their hearts. Control mice expressed wild-type RLC (WT-RLC), and HCM mice expressed the N47K RLC mutation. In the absence of mavacamten, skinned papillary muscle strips from WT-RLC mice produced greater isometric force than strips from N47K mice. Adding 0.3 µM mavacamten decreased maximal isometric force and reduced Ca2+ sensitivity of contraction for both genotypes, but this reduction in pCa50 was nearly twice as large for WT-RLC versus N47K. We also used stochastic length-perturbation analysis to characterize cross-bridge kinetics. The cross-bridge detachment rate was measured as a function of [MgATP] to determine the effect of mavacamten on myosin nucleotide handling rates. Mavacamten increased the MgADP release and MgATP binding rates for both genotypes, thereby contributing to faster cross-bridge detachment, which could speed up myocardial relaxation during diastole. Our data suggest that mavacamten reduces isometric tension and Ca2+ sensitivity of contraction via decreased strong cross-bridge binding. Mavacamten may become a useful therapy for patients with heart disease, including some forms of HCM.NEW & NOTEWORTHY Mavacamten is a pharmaceutical that binds to myosin, and it is under investigation as a therapy for some forms of heart disease. We show that mavacamten reduces isometric tension and Ca2+ sensitivity of contraction in skinned myocardial strips from a mouse model of hypertrophic cardiomyopathy that expresses the N47K mutation in cardiac myosin regulatory light chain. Mavacamten reduces contractility by decreasing strong cross-bridge binding, partially due to faster cross-bridge nucleotide handling rates that speed up myosin detachment.


Asunto(s)
Bencilaminas/farmacología , Señalización del Calcio/efectos de los fármacos , Cardiomiopatía Hipertrófica/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Contracción Miocárdica/efectos de los fármacos , Cadenas Ligeras de Miosina/metabolismo , Músculos Papilares/efectos de los fármacos , Uracilo/análogos & derivados , Miosinas Ventriculares/antagonistas & inhibidores , Animales , Cardiomiopatía Hipertrófica/enzimología , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/fisiopatología , Modelos Animales de Enfermedad , Humanos , Cinética , Masculino , Ratones Transgénicos , Mutación , Cadenas Ligeras de Miosina/genética , Músculos Papilares/enzimología , Músculos Papilares/fisiopatología , Uracilo/farmacología , Miosinas Ventriculares/metabolismo
6.
J Cell Mol Med ; 24(22): 13151-13162, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32989924

RESUMEN

Cardiac hypertrophy is a common pathological change in patients with progressive cardiac function failure, which can be caused by hypertrophic cardiomyopathy (HCM), dilated cardiomyopathy (DCM) or arterial hypertension. Despite years of study, there is still limited knowledge about the underlying molecular mechanisms for cardiac hypertrophy. NDUFA7, a subunit of NADH:ubiquinone oxidoreductase (complex I), has been reported to be a novel HCM associated gene. However, the biological role of NDUFA7 in heart remains unknown. In this study, we found that NDUFA7 exhibited high expression in the heart, and its level was significantly decreased in mice model of cardiac hypertrophy. Moreover, we demonstrated that ndufa7 knockdown in developing zebrafish embryos resulted in cardiac development and functional defects, associated with increased expression of pathological hypertrophy biomarkers nppa (ANP) and nppb (BNP). Mechanistic study demonstrated that ndufa7 depletion promoted ROS production and calcineurin signalling activation. Moreover, NDUFA7 depletion contributed to cardiac cell hypertrophy. Together, these results report for the first time that ndufa7 is implicated in pathological cardiac hypertrophy.


Asunto(s)
Cardiomegalia/patología , Cardiomiopatía Hipertrófica/patología , Complejo I de Transporte de Electrón/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Arterias/metabolismo , Biomarcadores/metabolismo , Calcineurina/metabolismo , Cardiomegalia/enzimología , Cardiomiopatía Hipertrófica/enzimología , Línea Celular , Modelos Animales de Enfermedad , Complejo I de Transporte de Electrón/genética , Técnicas de Silenciamiento del Gen , Genotipo , Corazón/crecimiento & desarrollo , Corazón/fisiopatología , Insuficiencia Cardíaca/metabolismo , Hipertensión/metabolismo , Ratones , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Distribución Tisular , Pez Cebra/embriología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
7.
BMC Vet Res ; 16(1): 26, 2020 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-32000761

RESUMEN

BACKGROUND: Hypertrophic cardiomyopathy is the most common cardiovascular cause of death in cats. Although the majority of cats remain asymptomatic, some may develop signs of chronic heart failure due to diastolic failure, arterial thromboembolism (ATE) or sudden cardiac death. Therefore, it is crucial to identify individuals that are in high risk of developing cardiac complications before the onset of life-threatening signs. Oxidative stress is the imbalance between the production and neutralisation of reactive oxygen species. Uncontrolled reactive oxygen species overproduction leads to protein and lipid peroxidation and damages the DNA strands, injuring the cells and leading to their death. The aim of the study was to evaluate the oxidative state in cats with hypertrophic cardiomyopathy and healthy controls. RESULTS: In total, 30 cats divided into three groups were assessed: animals with clinically evident hypertrophic cardiomyopathy (HCM; n = 8), subclinical hypertrophic cardiomyopathy (SUB-HCM; n = 11) and healthy controls (n = 11). The activity of superoxide dismutase was statistically significantly lower in animals with symptomatic and asymptomatic hypertrophic cardiomyopathy (HCM 0.99 ± 0.35 U/mL; SUB-HCM 1.39 ± 0.4 U/mL) compared to healthy cats (2.07 ± 0.76 U/mL, p < 0.01). The activity of catalase was significantly lower in the SUB-HCM group (19.4 ± 4.2 nmol/min/mL) compared to the HCM (23.6 ± 5.9 nmol/min/mL) and the control (30 ± 7.5 nmol/min/mL, p < 0.01) group. The activity of glutathione peroxidase was 4196 ± 353 nmol/min/mL in the HCM group, 4331 ± 451 nmol/min/mL in the SUB-HCM group and 4037 ± 341 nmol/min/mL in the control group and did not differ significantly between groups. The total antioxidant capacity of plasma was 602 ± 65.5 copper reducing equivalents (CRE) in the HCM group, 605.9 ± 39.9 CRE in the SUB-HCM group and 629 ± 77.5 CRE in the healthy cats and did not differ significantly between the groups. CONCLUSIONS: Activities of superoxide dismutase and catalase differed in cats with hypertrophic cardiomyopathy, however the activity of the latter was only significantly lower in asymptomatic stage of the disease. The potentially beneficial effect of antioxidative substances on the disease progression in the asymptomatic and symptomatic stage of this disease should also be examined.


Asunto(s)
Antioxidantes/análisis , Cardiomiopatía Hipertrófica/veterinaria , Enfermedades de los Gatos/patología , Estrés Oxidativo , Animales , Biomarcadores/sangre , Cardiomiopatía Hipertrófica/enzimología , Enfermedades de los Gatos/enzimología , Catalasa/sangre , Gatos , Estudios Transversales , Ecocardiografía/veterinaria , Femenino , Masculino , Proyectos Piloto , Superóxido Dismutasa/sangre
8.
Am J Med Genet A ; 179(7): 1235-1240, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31074094

RESUMEN

Cardiomyopathies are clinically heterogeneous disorders and are the leading cause of cardiovascular morbidity and mortality. Different etiologies have a significant impact on prognosis. Recently, novel biallelic loss-of-function pathogenic variants in alpha-kinase 3 (ALPK3) were implicated in causing early-onset pediatric cardiomyopathy (cardiomyopathy, familial hypertrophic 27; OMIM 618052). To date, eight patients, all presented during early childhood, were reported with biallelic ALPK3 pathogenic variants. We describe the molecular and clinical phenotype characterization of familial cardiomyopathy on one family with six affected individuals. We identified homozygosity for an ALPK3 deleterious sequence variant (NM_020778.4:c.639G>A:p.Trp213*) in all the affected individuals. They presented with either dilated cardiomyopathy that progressed to hypertrophic cardiomyopathy (HCM) or HCM with left ventricular noncompaction. The age of presentation in our cohort extends between infancy to the fourth decade. The phenotypic severity decreases with the progression of age.


Asunto(s)
Cardiomiopatía Dilatada/genética , Cardiomiopatía Hipertrófica/genética , Proteínas Musculares/genética , Mutación , Fenotipo , Proteínas Quinasas/genética , Adulto , Edad de Inicio , Secuencia de Bases , Cardiomiopatía Dilatada/diagnóstico , Cardiomiopatía Dilatada/enzimología , Cardiomiopatía Dilatada/fisiopatología , Cardiomiopatía Hipertrófica/diagnóstico , Cardiomiopatía Hipertrófica/enzimología , Cardiomiopatía Hipertrófica/fisiopatología , Niño , Preescolar , Consanguinidad , Femenino , Expresión Génica , Homocigoto , Humanos , Lactante , Masculino , Proteínas Musculares/metabolismo , Linaje , Proteínas Quinasas/metabolismo , Secuenciación del Exoma
9.
J Am Heart Assoc ; 7(2)2018 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-29358191

RESUMEN

BACKGROUND: Severe cardiac hypertrophy can lead to cardiac remodeling and even heart failure in the end, which is a leading cause of cardiovascular disease-related mortality worldwide. A disintegrin and metalloprotease-22 (ADAM22), a member of the transmembrane and secreted metalloendopeptidase family, participates in many biological processes, including those in the cardiovascular system. However, there is no explicit information on whether ADAM22 can regulate the process of cardiac hypertrophy; the effects that ADAM22 exerts in cardiac hypertrophy remain elusive. METHODS AND RESULTS: We observed significantly increased ADAM22 expression in failing hearts from patients with dilated cardiomyopathy and hypertrophic cardiomyopathy; the same trend was observed in mice induced by transaortic constriction and in neonatal rat cardiomyocytes treated by angiotensin II. Therefore, we constructed both cardiac-specific ADAM22 overexpression and knockout mice. At 4 weeks after transaortic constriction, cardiac-specific ADAM22 knockout, by the CRISPR/Cas9 (clustered regularly interspaced palindromic repeat (CRISPR)-Cas9) system, deteriorated the severity of cardiac hypertrophy in mice, whereas cardiac-specific ADAM22 overexpression mitigated the degrees of cardiac hypertrophy in mice. Similarly, altered ADAM22 expression modulated the angiotensin II-mediated cardiomyocyte hypertrophy in neonatal rat cardiomyocytes. After screening several signaling pathways, we found ADAM22 played a role in inhibition of protein kinase B (AKT) activation. Under the cardiac-specific ADAM22 knockout background, AKT activation was enhanced in transaortic constriction-induced mice and angiotensin II-stimulated neonatal rat cardiomyocytes, with a severe degree of cardiac hypertrophy. Treatment of a specific AKT inhibitor attenuated the transaortic constriction-enhanced AKT activation and cardiac hypertrophy in mice. CONCLUSIONS: The findings demonstrated that ADAM22 negatively regulates the AKT activation and the process of cardiac hypertrophy and may provide new insights into the pathobiological features of cardiac hypertrophy.


Asunto(s)
Proteínas ADAM/metabolismo , Hipertrofia Ventricular Izquierda/prevención & control , Miocitos Cardíacos/enzimología , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Función Ventricular Izquierda , Remodelación Ventricular , Proteínas ADAM/deficiencia , Proteínas ADAM/genética , Animales , Animales Recién Nacidos , Cardiomiopatía Dilatada/enzimología , Cardiomiopatía Dilatada/fisiopatología , Cardiomiopatía Hipertrófica/enzimología , Cardiomiopatía Hipertrófica/fisiopatología , Estudios de Casos y Controles , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Hipertrofia Ventricular Izquierda/enzimología , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/patología , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Ratas Sprague-Dawley , Transducción de Señal
10.
Am J Physiol Heart Circ Physiol ; 313(2): H283-H292, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28550180

RESUMEN

PRKAG2 encodes the γ2-subunit isoform of 5'-AMP-activated protein kinase (AMPK), a heterotrimeric enzyme with major roles in the regulation of energy metabolism in response to cellular stress. Mutations in PRKAG2 have been implicated in a unique hypertrophic cardiomyopathy (HCM) characterized by cardiac glycogen overload, ventricular preexcitation, and hypertrophy. We identified a novel, de novo PRKAG2 mutation (K475E) in a neonate with prenatal onset of HCM. We aimed to investigate the cellular impact, signaling pathways involved, and therapeutic options for K475E mutation using cells stably expressing human wild-type (WT) or the K475E mutant. In human embryonic kidney-293 cells, the K475E mutation induced a marked increase in the basal phosphorylation of T172 and AMPK activity, reduced sensitivity to AMP in allosteric activation, and a loss of response to phenformin. In H9c2 cardiomyocytes, the K475E mutation induced inhibition of AMPK and reduced the response to phenformin and increases in the phosphorylation of p70S6 kinase (p70S6K) and eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1). Primary fibroblasts from the patient with the K475E mutation also showed marked increases in the phosphorylation of p70S6K and 4E-BP1 compared with those from age-matched, nondiseased controls. Moreover, overexpression of K475E induced hypertrophy in H9c2 cells, which was effectively reversed by treatment with rapamycin. Taken together, we have identified a novel, de novo infantile-onset PRKAG2 mutation causing HCM. Our study suggests the K475E mutation induces alteration in basal AMPK activity and results in a hypertrophy phenotype involving the mechanistic target of rapamycin signaling pathway, which can be reversed with rapamycin.NEW & NOTEWORTHY We identified a novel, de novo PRKAG2 mutation (K475E) in the cystathionine ß-synthase 3 repeat, a region critical for AMP binding but with no previous reported mutation. Our data suggest the mutation affects AMP-activated protein kinase activity, activates cell growth pathways, and results in cardiac hypertrophy, which can be reversed with rapamycin.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Cardiomiopatía Hipertrófica/genética , Mutación Missense , Miocitos Cardíacos/enzimología , Transducción de Señal , Proteínas Quinasas Activadas por AMP/química , Proteínas Quinasas Activadas por AMP/metabolismo , Adenosina Monofosfato/metabolismo , Cardiomiopatía Hipertrófica/tratamiento farmacológico , Cardiomiopatía Hipertrófica/enzimología , Cardiomiopatía Hipertrófica/fisiopatología , Proteínas Portadoras/metabolismo , Estudios de Casos y Controles , Análisis Mutacional de ADN , Activación Enzimática , Fibroblastos/enzimología , Fibroblastos/patología , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Recién Nacido , Péptidos y Proteínas de Señalización Intracelular , Modelos Moleculares , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Fenformina/farmacología , Fenotipo , Fosfoproteínas/metabolismo , Fosforilación , Conformación Proteica , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Relación Estructura-Actividad , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Transfección
11.
Hum Pathol ; 49: 27-32, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26826406

RESUMEN

Complex I deficiency causes Leigh syndrome, fatal infant lactic acidosis, and neonatal cardiomyopathy. Mutations in more than 100 nuclear DNA and mitochondrial DNA genes miscode for complex I subunits or assembly factors. ACAD9 is an acyl-CoA dehydrogenase with a novel function in assembly of complex I; biallelic mutations cause progressive encephalomyopathy, recurrent Reye syndrome, and fatal cardiomyopathy. We describe the first autopsy in fatal neonatal lethal lactic acidosis due to mutations in ACAD9 that reduced complex I activity. We identified mitochondrial hyperplasia in cardiac myocytes, diaphragm muscle, and liver and renal tubules in formalin-fixed, paraffin-embedded tissue using immunohistochemistry for mitochondrial antigens. Whole-exome sequencing revealed compound heterozygous variants in the ACAD9 gene: c.187G>T (p.E63*) and c.941T>C (p.L314P). The nonsense mutation causes late infantile lethality; the missense variant is novel. Autopsy-derived fibroblasts had reduced complex I activity (53% of control) with normal activity in complexes II to IV, similar to reported cases of ACAD9 deficiency.


Asunto(s)
Acidosis Láctica/diagnóstico , Acidosis/diagnóstico , Acil-CoA Deshidrogenasa/deficiencia , Acil-CoA Deshidrogenasas/genética , Errores Innatos del Metabolismo de los Aminoácidos/diagnóstico , Cardiomiopatía Hipertrófica/diagnóstico , Codón sin Sentido , Diafragma/patología , Complejo I de Transporte de Electrón/deficiencia , Túbulos Renales/patología , Enfermedad de Leigh/diagnóstico , Mitocondrias Cardíacas/patología , Mitocondrias Hepáticas/patología , Mitocondrias Musculares/patología , Enfermedades Mitocondriales/diagnóstico , Insuficiencia Multiorgánica/diagnóstico , Debilidad Muscular/diagnóstico , Acidosis/enzimología , Acidosis/genética , Acidosis/patología , Acidosis Láctica/enzimología , Acidosis Láctica/genética , Acidosis Láctica/patología , Acil-CoA Deshidrogenasa/genética , Acil-CoA Deshidrogenasas/deficiencia , Errores Innatos del Metabolismo de los Aminoácidos/enzimología , Errores Innatos del Metabolismo de los Aminoácidos/genética , Errores Innatos del Metabolismo de los Aminoácidos/patología , Autopsia , Cardiomiopatía Hipertrófica/enzimología , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/patología , Causas de Muerte , Células Cultivadas , Análisis Mutacional de ADN , ADN Mitocondrial/genética , Diafragma/enzimología , Complejo I de Transporte de Electrón/genética , Resultado Fatal , Fibroblastos/enzimología , Fibroblastos/patología , Predisposición Genética a la Enfermedad , Humanos , Hiperplasia , Inmunohistoquímica , Recién Nacido , Túbulos Renales/enzimología , Enfermedad de Leigh/enzimología , Enfermedad de Leigh/genética , Enfermedad de Leigh/patología , Masculino , Mitocondrias Cardíacas/enzimología , Mitocondrias Hepáticas/enzimología , Mitocondrias Musculares/enzimología , Enfermedades Mitocondriales/enzimología , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/patología , Insuficiencia Multiorgánica/enzimología , Insuficiencia Multiorgánica/genética , Insuficiencia Multiorgánica/patología , Debilidad Muscular/enzimología , Debilidad Muscular/genética , Debilidad Muscular/patología , Fenotipo , Transfección
12.
J Cardiovasc Magn Reson ; 17: 89, 2015 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-26496977

RESUMEN

BACKGROUND: Autosomal dominantly inherited PRKAG2 cardiac syndrome is due to a unique defect of the cardiac cell metabolism and has a distinctive histopathology with excess intracellular glycogen, and prognosis different from sarcomeric hypertrophic cardiomyopathy. We aimed to define the distinct characteristics of PRKAG2 using cardiovascular magnetic resonance (CMR). METHODS: CMR (1.5 T) and genetic testing were performed in two families harboring PRKAG2 mutations. On CMR, segmental analysis of left ventricular (LV) hypertrophy (LVH), function, native T1 mapping, and late gadolinium enhancement (LGE) were performed. RESULTS: Six individuals (median age 23 years, range 16-48; two females) had a PRKAG2 mutation: five with an R302Q mutation (family 1), and one with a novel H344P mutation (family 2). Three of six mutation carriers had LV mass above age and gender limits (203 g/m2, 157 g/m2 and 68 g/m2) and others (with R302Q mutation) normal LV masses. All mutation carriers had LVH in at least one segment, with the median maximal wall thickness of 13 mm (range 11-37 mm). Two R302Q mutation carriers with markedly increased LV mass (203 g/m2 and 157 g/m2) showed a diffuse pattern of hypertrophy but predominantly in the interventricular septum, while other mutation carriers exhibited a non-symmetric mid-infero-lateral pattern of hypertrophy. In family 1, the mutation negative male had a mean T1 value of 963 ms, three males with the R302Q mutation, LVH and no LGE a mean value of 918 ± 11 ms, and the oldest male with the R302Q mutation, extensive hypertrophy and LGE a mean value of 973 ms. Of six mutations carriers, two with advanced disease had LGE with 11 and 22 % enhancement of total LV volume. CONCLUSIONS: PRKAG2 cardiac syndrome may present with eccentric distribution of LVH, involving focal mid-infero-lateral pattern in the early disease stage, and more diffuse pattern but focusing on interventricular septum in advanced cases. In patients at earlier stages of disease, without LGE, T1 values may be reduced, while in the advanced disease stage T1 mapping may result in higher values caused by fibrosis. CMR is a valuable tool in detecting diffuse and focal myocardial abnormalities in PRKAG2 cardiomyopathy.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/patología , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/patología , Imagen por Resonancia Cinemagnética , Mutación , Miocardio/patología , Adolescente , Adulto , Cardiomiopatía Hipertrófica/enzimología , Cardiomiopatía Hipertrófica/fisiopatología , Medios de Contraste , Análisis Mutacional de ADN , Electrocardiografía , Femenino , Fibrosis , Predisposición Genética a la Enfermedad , Humanos , Hipertrofia Ventricular Izquierda/enzimología , Hipertrofia Ventricular Izquierda/fisiopatología , Masculino , Meglumina , Persona de Mediana Edad , Compuestos Organometálicos , Fenotipo , Valor Predictivo de las Pruebas , Función Ventricular Izquierda , Remodelación Ventricular , Adulto Joven
13.
Int J Cardiovasc Imaging ; 31(4): 669-79, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25627778

RESUMEN

Consistent protocols for the assessment of diastolic and systolic cardiac function to assure the comparability of existing data on preclinical models are missing. Calcineurin transgene (CN) mice are a preclinical model for hypertrophic and failing hearts. We aimed at evaluating left and right ventricular structural and functional remodeling in CN hearts with an optimized phenotyping protocol. We developed a protocol using techniques and indices comparable to those from human diagnostics for comprehensive in vivo cardiac screening using high-frequency echocardiography, Doppler, electrocardiography and cardiac magnetic resonance (CMR) techniques. We measured left and right ventricular dimensions and function, pulmonary and mitral flow pattern and the hearts electrophysiology non-invasively in <1 h per mouse. We found severe biventricular dilation and a drastic decline in performance in accordance with a condition of heart failure (HF), diastolic dysfunction and defects in electrical conduction in 8-week-old calcineurin transgenic mice. Echocardiography of the left ventricle was performed with and without anesthesia. In all cases absolute values on echocardiography compared with CMR were smaller for LV dimension and wall thickness, resulting in higher fractional shorting and ejection fraction. The study protocol described here opens opportunities to assess the added value of combined echocardiography, Doppler, CMR and ECG recording techniques for the diagnosis of biventricular cardiac pathologies i.e. of HF and to study symptom occurrence and disease progression non-invasively in high-throughput. Phenotyping CN hearts revealed new symptom occurrence and allowed insights into the diverse phenotype of hypertrophic failing hearts.


Asunto(s)
Calcineurina/genética , Cardiomiopatía Hipertrófica/diagnóstico , Insuficiencia Cardíaca/diagnóstico , Ensayos Analíticos de Alto Rendimiento/métodos , Hipertrofia Ventricular Izquierda/diagnóstico , Hipertrofia Ventricular Derecha/diagnóstico , Disfunción Ventricular Izquierda/diagnóstico , Disfunción Ventricular Derecha/diagnóstico , Animales , Calcineurina/metabolismo , Cardiomiopatía Hipertrófica/enzimología , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/fisiopatología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ecocardiografía Doppler , Electrocardiografía , Femenino , Predisposición Genética a la Enfermedad , Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/fisiopatología , Hemodinámica , Hipertrofia Ventricular Izquierda/enzimología , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/fisiopatología , Hipertrofia Ventricular Derecha/enzimología , Hipertrofia Ventricular Derecha/genética , Hipertrofia Ventricular Derecha/fisiopatología , Imagen por Resonancia Magnética , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Cadenas Pesadas de Miosina/genética , Fenotipo , Valor Predictivo de las Pruebas , Regiones Promotoras Genéticas , Disfunción Ventricular Izquierda/enzimología , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/fisiopatología , Disfunción Ventricular Derecha/enzimología , Disfunción Ventricular Derecha/genética , Disfunción Ventricular Derecha/fisiopatología , Función Ventricular Izquierda , Función Ventricular Derecha , Miosinas Ventriculares/genética , Remodelación Ventricular
14.
J Mol Cell Cardiol ; 77: 53-63, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25280781

RESUMEN

The mechanisms linking the expression of sarcomeric mutant proteins to the development of pathological hypertrophy in hypertrophic cardiomyopathy (HCM) remain poorly understood. We investigated the role of the plasma membrane Ca(2+)-ATPase PMCA4 in the HCM phenotype using a transgenic model that expresses mutant (Glu180Gly) α-tropomyosin (Tm180) in heart. Immunoblot analysis revealed that cardiac PMCA4 expression was upregulated early in Tm180 disease pathogenesis. This was accompanied by an increase in levels of the L-type Ca(2+)-channel, which is implicated in pathological hypertrophy. When Tm180 mice were crossed with a PMCA4-null line, loss of PMCA4 caused the abrogation of hypertrophy in Tm180/PMCA4-null double mutant mice. RT-PCR analysis of Tm180/PMCA4-null hearts revealed blunting of the fetal program and reversion of pro-fibrotic Col1a1 and Col3a1 gene expression to wild-type levels. This was accompanied by evidence of reduced L-type Ca(2+)-channel expression, and diminished calcineurin activity. Expression of the metabolic substrate transporters glucose transporter 4 and carnitine palmitoyltransferase 1b was preserved and Tm180-related changes in mRNA levels of various contractile stress-related proteins including the cardiac ankyrin protein CARP and the N2B isoform of titin were reversed in Tm180/PMCA4-null hearts. cGMP levels were increased and phosphorylation of vasodilator-stimulated phosphoprotein was elevated in Tm180/PMCA4-null hearts. These changes were associated with a sharp reduction in left ventricular end-diastolic pressure in Tm180/PMCA4-null hearts, which occurred despite persistence of Tm180-related impairment of relaxation dynamics. These results reveal a novel and specific role for PMCA4 in the Tm180 hypertrophic phenotype, with the "protective" effects of PMCA4 deficiency encompassing multiple determinants of HCM-related hypertrophy.


Asunto(s)
Cardiomiopatía Hipertrófica/enzimología , ATPasas Transportadoras de Calcio de la Membrana Plasmática/genética , Tropomiosina/genética , Animales , Cardiomiopatía Hipertrófica/genética , Modelos Animales de Enfermedad , Expresión Génica , Técnicas de Inactivación de Genes , Frecuencia Cardíaca , Masculino , Ratones Noqueados , Miocardio/metabolismo , Miocardio/patología , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Tropomiosina/metabolismo , Presión Ventricular
15.
Nat Commun ; 5: 4241, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24980141

RESUMEN

The Rag family proteins are Ras-like small GTPases that have a critical role in amino-acid-stimulated mTORC1 activation by recruiting mTORC1 to lysosome. Despite progress in the mechanistic understanding of Rag GTPases in mTORC1 activation, little is known about the physiological function of Rag GTPases in vivo. Here we show that loss of RagA and RagB (RagA/B) in cardiomyocytes results in hypertrophic cardiomyopathy and phenocopies lysosomal storage diseases, although mTORC1 activity is not substantially impaired in vivo. We demonstrate that despite upregulation of lysosomal protein expression by constitutive activation of the transcription factor EB (TFEB) in RagA/B knockout mouse embryonic fibroblasts, lysosomal acidification is compromised owing to decreased v-ATPase level in the lysosome fraction. Our study uncovers RagA/B GTPases as key regulators of lysosomal function and cardiac protection.


Asunto(s)
Cardiomiopatía Hipertrófica/enzimología , Lisosomas/enzimología , Proteínas de Unión al GTP Monoméricas/metabolismo , Animales , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/prevención & control , Femenino , Humanos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Unión al GTP Monoméricas/genética , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Miocitos Cardíacos/enzimología , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
16.
J Intern Med ; 276(6): 543-59, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24824502

RESUMEN

The AMP-activated protein kinase (AMPK) is a sensor of cellular energy status that regulates cellular and whole-body energy balance. A recently reported crystal structure has illuminated the complex regulatory mechanisms by which AMP and ADP cause activation of AMPK, involving phosphorylation by the upstream kinase LKB1. Once activated by falling cellular energy status, AMPK activates catabolic pathways that generate ATP whilst inhibiting anabolic pathways and other cellular processes that consume ATP. A role of AMPK is implicated in many human diseases. Mutations in the γ2 subunit cause heart disease due to excessive glycogen storage in cardiac myocytes, leading to ventricular pre-excitation. AMPK-activating drugs reverse many of the metabolic defects associated with insulin resistance, and recent findings suggest that the insulin-sensitizing effects of the widely used antidiabetic drug metformin are mediated by AMPK. The upstream kinase LKB1 is a tumour suppressor, and AMPK may exert many of its antitumour effects. AMPK activation promotes the oxidative metabolism typical of quiescent cells, rather than the aerobic glycolysis observed in tumour cells and cells involved in inflammation, explaining in part why AMPK activators have both antitumour and anti-inflammatory effects. Salicylate (the major in vivo metabolite of aspirin) activates AMPK, and this could be responsible for at least some of the anticancer and anti-inflammatory effects of aspirin. In addition to metformin and salicylates, novel drugs that modulate AMPK are likely to enter clinical trials soon. Finally, AMPK may be involved in viral infection: downregulation of AMPK during hepatitis C virus infection appears to be essential for efficient viral replication.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Metabolismo Energético , Proteínas Quinasas Activadas por AMP/química , Proteínas Quinasas Activadas por AMP/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/genética , Nucleótidos de Adenina/metabolismo , Canales de Calcio/metabolismo , Cardiomiopatía Hipertrófica/enzimología , Diabetes Mellitus Tipo 2/enzimología , Humanos , Inflamación/enzimología , Estructura Molecular , Mutación , Neoplasias/enzimología , Virosis/enzimología
17.
PLoS One ; 8(5): e63309, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23691019

RESUMEN

BACKGROUND: Genetic factors in the pathogenesis of cardiomyopathies have received a lot attention during the past two decades. Angiotensin I converting enzyme (ACE) insertion/deletion (I/D) polymorphisms were found to be associated with cardiomyopathies. However, the previous results were inconsistent. The current meta-analysis aims to examine the association of ACE I/D polymorphisms and dilated cardiomyopathy (DCM) or hypertrophic cardiomyopathy (HCM). METHODS: Eight studies on DCM (1387 controls and 977 patients) and eight studies on HCM (1055 controls and 827 patients) were included in this meta-analysis. RESULTS: The overall data showed no significant association between ACE I/D polymorphism and DCM risk. Further subgroup analysis by ethnicity also did not find a significantly increased risk for D allele carriers among East Asians and Europeans. However, the overall analysis suggested that the D allele carriers might be associated with increased risk of HCM (DD/ID vs. II: OR = 1.69, 95% CI 1.04-2.74, P = 0.03). CONCLUSION: In summary, the meta-analysis indicated that certain ACE I/D polymorphism might be associated with HCM but not DCM susceptibility. Given the limited sample sizes, further large multicenter case-control investigation is needed.


Asunto(s)
Cardiomiopatía Dilatada/genética , Cardiomiopatía Hipertrófica/genética , Predisposición Genética a la Enfermedad/genética , Mutación INDEL , Peptidil-Dipeptidasa A/genética , Polimorfismo Genético , Cardiomiopatía Dilatada/enzimología , Cardiomiopatía Hipertrófica/enzimología , Humanos
18.
Arch Biochem Biophys ; 535(1): 39-48, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23352598

RESUMEN

The pathological progression of hypertrophic cardiomyopathy (HCM) is sexually dimorphic such that male HCM mice develop phenotypic indicators of cardiac disease well before female HCM mice. Here, we hypothesized that alterations in myofilament function underlies, in part, this sex dimorphism in HCM disease development. Firstly, 10-12month female HCM (harboring a mutant [R403Q] myosin heavy chain) mice presented with proportionately larger hearts than male HCM mice. Next, we determined Ca(2+)-sensitive tension development in demembranated cardiac trabeculae excised from 10-12month female and male HCM mice. Whereas HCM did not impact Ca(2+)-sensitive tension development in male trabeculae, female HCM trabeculae were more sensitive to Ca(2+) than wild-type (WT) counterparts and both WT and HCM males. We hypothesized that the underlying cause of this sex difference in Ca(2+)-sensitive tension development was due to changes in Ca(2+) handling and sarcomeric proteins, including expression of SR Ca(2+) ATPase (2a) (SERCA2a), ß-myosin heavy chain (ß-MyHC) and post-translational modifications of myofilament proteins. Female HCM hearts showed an elevation of SERCA2a and ß-MyHC protein whereas male HCM hearts showed a similar elevation of ß-MyHC protein but a reduced level of cardiac troponin T (cTnT) phosphorylation. We also measured the distribution of cardiac troponin I (cTnI) phosphospecies using phosphate-affinity SDS-PAGE. The distribution of cTnI phosphospecies depended on sex and HCM. In conclusion, female and male HCM mice display sex dimorphic myofilament function that is accompanied by a sex- and HCM-dependent distribution of sarcomeric proteins and cTnI phosphospecies.


Asunto(s)
Cardiomiopatía Hipertrófica/metabolismo , Miofibrillas/fisiología , Troponina I/metabolismo , Animales , Calcio/metabolismo , Cardiomiopatía Hipertrófica/enzimología , Cardiomiopatía Hipertrófica/patología , Electroforesis en Gel de Poliacrilamida , Femenino , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Masculino , Ratones , Tono Muscular , Mutación , Miofibrillas/genética , Miofibrillas/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Factores Sexuales , Troponina T/metabolismo , Miosinas Ventriculares/genética , Miosinas Ventriculares/metabolismo
19.
Cardiovasc Res ; 97(1): 44-54, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22987565

RESUMEN

AIMS: The role of calcineurin protein phosphatase 2B (PP2B) in the pathogenesis of human hypertrophic cardiomyopathy (HCM) remains unsettled. We determined potential involvement of calcineurin in the pathogenesis of HCM caused by mutations in myozenin 2 (MYOZ2), an inhibitor of calcineurin. METHODS AND RESULTS: We generated multiple lines of transgenic mice expressing either Flag-tagged wild-type (WT) (MYOZ2(WT)) or mutant MYOZ2(S48P) and MYOZ2(I246M), identified in families with HCM, in the heart. To mimic the human genotype, we generated bigenic mice expressing WT and mutant MYOZ2 in the background of hemizygous endogenous MYOZ2 (Myoz2(+/-)). Transgene proteins constituted 15-48% of the total MYOZ2 protein in the heart. Mutant MYOZ2 mice showed molecular, cellular, and gross cardiac hypertrophy, preserved systolic function, and interstitial fibrosis. Immunofluorescence staining showed co-localization of WT and mutant MYOZ2 proteins with α-actinin at the Z disks. Electron microscopy showed disrupted and mal-aligned Z disks in the mutant mice. Cardiac calcineurin activity, determined by quantifying Rcan1.4 mRNA and protein levels, luciferase activity in triple transgenic Myoz2(+/-):NFATc-Luc:MYOZ2(I246M) and Myoz2(+/-):NFATc-Luc:MYOZ2(WT) mice, and NFATc transcriptional activity assay, was unchanged in the mutant transgenic mice. However, levels of phospho-ERK1/2 and JNK54/46 were altered in the transgenic mice. Likewise, lentiviral-mediated expression of the MYOZ2(I246M) did not affect RCAN1.4 and calcineurin (PPP3CB) protein levels. CONCLUSIONS: Thus, the cardiac phenotype in HCM caused by MYOZ2 mutations might be independent of calcineurin activity in the heart. Z disk abnormalities might provide the stimulus for the induction of cardiac hypertrophy caused by MYOZ2 mutations.


Asunto(s)
Calcineurina/metabolismo , Cardiomiopatía Hipertrófica/genética , Proteínas Portadoras/genética , Proteínas Musculares/genética , Mutación , Miocardio/enzimología , Animales , Proteínas de Unión al Calcio , Cardiomiopatía Hipertrófica/enzimología , Cardiomiopatía Hipertrófica/patología , Cardiomiopatía Hipertrófica/fisiopatología , Proteínas Portadoras/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibrosis , Técnica del Anticuerpo Fluorescente , Genes Reporteros , Predisposición Genética a la Enfermedad , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas de Microfilamentos , Microscopía Electrónica , Proteínas Musculares/deficiencia , Proteínas Musculares/metabolismo , Miocardio/ultraestructura , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Fenotipo , Fosforilación , ARN Mensajero/metabolismo , Ratas , Transfección
20.
Rev. chil. cardiol ; 32(3): 233-239, 2013. ilus
Artículo en Español | LILACS | ID: lil-705227

RESUMEN

El Síndrome de Sengers es una enfermedad mitocondrial autosómica recesiva, producida por mutación del gen de la Acil-Glicerol Kinasa (AGK), recientemente descubierto. Se caracteriza por cataratas congénitas bilaterales, miocardiopatía hipertrófica y acidosis láctica. Puede tener miopatía esquelética leve, intolerancia al ejercicio y desarrollo mental normal. Los pacientes fallecen tempranamente debido a falla cardíaca. Dada la alta letalidad, lo infrecuente de este síndrome y la presencia de un diagnóstico confirmado, se presenta el caso clínico de 2 hermanos chilenos, fallecidos por la enfermedad, que se presentaron con el cuadro característico de cataratas congénitas bilaterales, miocardiopa-tía hipertrófica y acidosis láctica. El mayor, se operó las cataratas a los 4 meses de edad y falleció a la edad de 13 meses debido a falla cardíaca severa refractaria y falla orgánica múltiple, descompensado por una infección respiratoria. El menor se diagnosticó a los 3 meses de edad y se le confirmó la mutación del gen de AGK en Alemania. Se decidió no operarlo de las cataratas dado el mal pronóstico vital. Presentó progresión de la miocardiopatía hipertrófica y falleció súbitamente a los 8 meses de edad.


Senger's Syndrome is a recessive autosomal mitochondrial disease due to a recently discovered mutation of the Acyl-Glycerol Kinase (AGK) gen,. It is characterized by congenital bilateral cataracts, progressive hypertrophic cardiomyopathy and lactic acidosis. It may present skeletal myopathy, exercise intolerance and usually normal mental development. Patients die early in life due to heart failure. The clinical cases of two brothers with a confirmed diagnosis of Senger's syndrome are reported. The older brother was operated on for cataracts at the age of 4 months and he died when he was 13 months old due to severe refractory heart failure and multi-organ failure, decompensated by a respiratory infection. The younger brother was diagnosed at 3 month of age and the AGK gene mutation was confirmed in Germany. Cataracts were not operated on due to the the patient's extremely poor prognosis. He had progressive hypertrophic cardiomyopathy and died suddenly at 8 month of age.


Asunto(s)
Humanos , Masculino , Recién Nacido , Lactante , Cardiomiopatía Hipertrófica/enzimología , Cardiomiopatía Hipertrófica/genética , Enfermedades Mitocondriales , Fosfotransferasas (Aceptor de Grupo Alcohol) , Acidosis Láctica , Cardiomiopatías , Catarata/congénito , Mutación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...