Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Lab Invest ; 100(6): 900, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32203148

RESUMEN

This article was originally published under Nature Research's License to Publish, but has now been made available under a [CC BY 4.0] license. The PDF and HTML versions of the article have been modified accordingly.

2.
Lab Invest ; 100(6): 887-899, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32060408

RESUMEN

Genetic cardiomyopathy is a group of intractable cardiovascular disorders involving heterogeneous genetic contribution. This heterogeneity has hindered the development of life-saving therapies for this serious disease. Genetic mutations in dystrophin and its associated glycoproteins cause cardiomuscular dysfunction. Large animal models incorporating these genetic defects are crucial for developing effective medical treatments, such as tissue regeneration and gene therapy. In the present study, we knocked out the δ-sarcoglycan (δ-SG) gene (SGCD) in domestic pig by using a combination of efficient de novo gene editing and somatic cell nuclear transfer. Loss of δ-SG expression in the SGCD knockout pigs caused a concomitant reduction in the levels of α-, ß-, and γ-SG in the cardiac and skeletal sarcolemma, resulting in systolic dysfunction, myocardial tissue degeneration, and sudden death. These animals exhibited symptoms resembling human genetic cardiomyopathy and are thus promising for use in preclinical studies of next-generation therapies.


Asunto(s)
Cardiomiopatías , Sarcoglicanos , Animales , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Femenino , Mutación del Sistema de Lectura/genética , Técnicas de Inactivación de Genes , Masculino , Miocardio/química , Miocardio/metabolismo , Miocardio/patología , Sarcoglicanos/deficiencia , Sarcoglicanos/genética , Porcinos
3.
J Hum Genet ; 64(4): 351-353, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30631120

RESUMEN

Since mitochondria are energy-generating micro-organisms, most of the disorders in patients with mitochondrial diseases (mt-disease) are considered secondary to defects in ATP synthesis, although some other factors such as reactive oxygen species may be involved. A simultaneous oral administration of febuxostat and inosine was reported to elevate both hypoxanthine and ATP levels in peripheral blood. Based on those results, we attempted co-administration of febuxostat and inosine in two patients with mitochondrial disease: one patient with mitochondrial cardiomyopathy and the other patient with mitochondrial diabetes. In the former case, brain natriuretic peptide (BNP), which is a specific marker for heart failure, was decreased by 31%, and in the latter case, the insulinogenic index increased 3.1 times, suggesting the favorable action of the treatment. Considering that there is no effective treatment available for this disorder, the present therapy may be quite useful for the management of patients with mitochondrial diseases.


Asunto(s)
Adenosina Trifosfato/biosíntesis , Mitocondrias/metabolismo , Enfermedades Mitocondriales/tratamiento farmacológico , Péptido Natriurético Encefálico/metabolismo , Anciano de 80 o más Años , Cardiomiopatías/tratamiento farmacológico , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Febuxostat/administración & dosificación , Femenino , Humanos , Hipoxantina/metabolismo , Inosina/administración & dosificación , Masculino , Persona de Mediana Edad , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/patología , Especies Reactivas de Oxígeno/metabolismo
4.
Mol Cell Biochem ; 323(1-2): 149-59, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19083155

RESUMEN

The precise role of delta-sarcoglycan (SG) that is constitutively expressed in skeletal muscle cells and may serve for maintaining the sarcolemmal integrity has not been identified. The delta-SG protein is at first among SG complex. To specifically identify the role in C(2)C(12) cells during the myogenesis, we screened several RNA interference (RNAi) candidates at first, and knocked down both levels of the mRNA and protein, employing adenovirus-mediated RNAi. We found no morphological alteration at both myoblast and myotube stages by suppression of delta-SG. The specific knockdown of delta-SG accompanied a concomitant decrease of alpha-, beta-, and gamma-SGs preserving normal levels of each transcript. As for the localization, alpha-, beta-, and gamma-SGs were weakly stained on the cell membrane in delta-SG knockdown cells, whereas each SG in control cell was localized both on the cell membrane and myoplasm abundantly. This enhanced post-translational loss would represent similitude of the progression of cardiomuscular diseases in vitro. Different from cardiac muscle cells, skeletal muscle cell culture without muscle contraction may imply that mechanical stress per se is not primarily involved in the progression of limb-girdle muscular dystrophy. Furthermore, we have observed translocation of calpain-2 to cell membrane in delta-SG knockdown cells, suggesting that Ca(2+)-sensitive proteases, calpains closely take part in post-translational proteolysis.


Asunto(s)
Isoformas de Proteínas/metabolismo , Sarcoglicanos/genética , Animales , Calpaína/metabolismo , Línea Celular , Humanos , Ratones , Ratones Noqueados , Desarrollo de Músculos/fisiología , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/patología , Distrofia Muscular de Cinturas/fisiopatología , Isoformas de Proteínas/genética , Interferencia de ARN , Sarcoglicanos/metabolismo , Estrés Mecánico
5.
Ann Allergy Asthma Immunol ; 101(2): 153-9, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18727470

RESUMEN

BACKGROUND: Although historical support exists for the concept that breastfeeding might be protective against allergic diseases, contradictory findings have been observed recently. OBJECTIVE: To investigate the cumulative prevalence of allergic diseases in Japanese university students and to identify explanatory variables including breastfeeding. METHODS: From March 18, 2003, through March 29, 2005, a total of 9,615 students newly enrolled at the University of Tokyo responded to a written questionnaire on allergic diseases. RESULTS: Cumulative prevalence of allergic rhinitis, atopic dermatitis, and asthma was 47.2%, 17.4%, and 9.3%, respectively. These data were closely correlated, and prevalence of any 1 of the 3 diseases significantly increased the odds for historical prevalence of the other 2 (P < .001). Male sex (odds ratio [OR], 1.5; 95% confidence interval [CI], 1.4-1.7) and maternal (OR, 2.2; 95% CI, 2.0-2.5) or paternal (OR, 1.6; 95% CI, 1.4-1.8) history of allergic rhinitis were significant correlates of increased odds for allergic rhinitis. Maternal (OR, 2.7; 95% CI, 1.6-4.5), paternal (OR, 3.8; 95% CI, 2.2-6.6), or sibling (OR, 1.9; 95% CI, 1.5-2.4) history of atopic dermatitis was a significant correlate of increased odds for atopic dermatitis. As for asthma, maternal (OR, 4.9; 95% CI, 3.0-7.9), paternal (OR, 4.0; 95% CI, 2.3-7.0), or sibling (OR, 3.3; 95% CI, 2.4-4.5) history of asthma was a significant correlate of increased odds. Logistic regression analysis showed no consistent evidence of the effects of breastfeeding on the cumulative prevalence. CONCLUSION: The cumulative prevalence of these diseases among young adults revealed that the effect of breastfeeding is negligible when compared with genetic factors.


Asunto(s)
Lactancia Materna , Dermatitis Atópica/epidemiología , Hipersensibilidad Respiratoria/epidemiología , Adolescente , Adulto , Asma/epidemiología , Femenino , Encuestas Epidemiológicas , Humanos , Japón/epidemiología , Modelos Logísticos , Masculino , Prevalencia , Rinitis Alérgica Perenne/epidemiología , Rinitis Alérgica Estacional/epidemiología , Encuestas y Cuestionarios
6.
Int Heart J ; 49(1): 49-58, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18360064

RESUMEN

HMG-CoA reductase inhibitors (statins) have recently been reported to improve cardiac function, and decrease the incidence of heart failure (HF) in hyperlipidemic patients. However, evidence for statin treatment in patients with HF remains a subject of debate. Thus, a study was initiated to examine the effects of pitavastatin on cardiac function evaluated by echocardiographic findings and plasma brain natriuretic peptide (BNP) levels in patients with HF. Twenty-three patients with HF were treated with pitavastatin 1-2 mg/day in addition to standard therapy for 7.5 +/- 3.8 months. Left ventricular end-diastolic dimension (LVDd) and left ventricular end-systolic dimension (LVDs) were determined by echocardiography. Left ventricular ejection fraction (LVEF) was calculated using Teichholz's formula. Serum lipid and plasma BNP levels were also measured. During the follow-up period, LVEF was increased from 42 +/- 11 to 48 +/- 13% (P = 0.002). LVDs was reduced from 43 +/- 10 to 40 +/- 10 mm (P < 0.001), while there was no change in LVDd. E/A (n = 10) and deceleration time (n = 7), obtained in some patients, did not change significantly (0.89 +/- 0.33 to 0.77 +/- 0.17%, and 215 +/- 46 to 227 +/- 72 msec, respectively). In addition, the plasma BNP level was moderately, but significantly decreased from 94 +/- 78 to 70 +/- 56 pg/mL (P = 0.005). In subgroup analysis, LVEF was improved in both patients with ischemic and nonischemic HF. There was no significant correlation between the percent change in serum total cholesterol and the percent change in LVEF by pitavastatin treatment. Serum total cholesterol, LDL-cholesterol, and triglycerides decreased by 21%, 30%, and 15%, respectively, and HDL-cholesterol increased by 12%. Pitavastatin improved cardiac function in patients with HF, which generally worsens with time. The results suggest that pitavastatin may be beneficial for treatment of HF.


Asunto(s)
Ecocardiografía , Inhibidores Enzimáticos/farmacología , Insuficiencia Cardíaca/fisiopatología , Péptido Natriurético Encefálico/sangre , Quinolinas/farmacología , Anciano , Anciano de 80 o más Años , Inhibidores Enzimáticos/uso terapéutico , Femenino , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/prevención & control , Hemodinámica/efectos de los fármacos , Humanos , Lípidos/sangre , Masculino , Persona de Mediana Edad , Quinolinas/uso terapéutico , Volumen Sistólico/efectos de los fármacos , Función Ventricular Izquierda/efectos de los fármacos
7.
Am J Physiol Cell Physiol ; 294(4): C957-65, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18216163

RESUMEN

Fusion of mononuclear myoblast to multinucleated myotubes is crucial for myogenesis. Both mu- and m-calpain are ubiquitously expressed in most cells and are particularly abundant in muscle cells. Knockout of calpain-1 (catalytic subunit of mu-calpain) induced moderate platelet dysaggregation, preserving the normal development and growth, although knockout of calpain-2 (m-calpain) is lethal in mice. Therefore, there should be muscle-specific function of m-calpain per se. Previous methods lack direct evidence for the involvement of m-calpain, because the specific inhibitor to m-calpain has not been developed yet and the inhibition was less potent. Here, we show that screened RNA interference (RNAi) specifically blocked the m-calpain expression by 95% at both the protein and the activity levels. After transfection of adenovirus vector-mediated cDNA corresponding to the RNAi-induced short hairpin RNA, m-calpain in C(2)C(12) myoblasts was knocked down with no compensatory overexpression of mu-calpain or calpain-3. The specific knockdown strongly inhibited the fusion to multinucleated myotubes. In addition, the knockdown modestly blocked ubiquitous effects, including cell migration, cell spreading, and alignment of central stress fiberlike structures. These results may indicate that m-calpain requiring millimolar Ca(2+) level for the full activation plays specific roles in myogenesis, independent of mu-calpain, and leave us challenging problems in the future.


Asunto(s)
Calpaína/metabolismo , Desarrollo de Músculos/fisiología , Mioblastos/metabolismo , Interferencia de ARN , Animales , Calpaína/genética , Diferenciación Celular , Fusión Celular , Línea Celular , Movimiento Celular , ADN Complementario , Regulación de la Expresión Génica/fisiología , Ratones , Desarrollo de Músculos/genética , Mioblastos/citología , ARN Mensajero/genética , ARN Mensajero/metabolismo
8.
Biochem Biophys Res Commun ; 369(1): 270-6, 2008 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-18047831

RESUMEN

Clinical efficacy of skeletal myoblast (skMb) transplantation is controversial whether this treatment produces beneficial outcome in patients with dilated cardiomyopathy (DCM). Based on immunological tolerance between wild-type and DCM hamsters with the deletion of delta-sarcoglycan (SG) gene, skMb engraftment in TO-2 myocardium (3x10(5) cells in approximately 100mg heart) was verified by the donor-specific expression of delta-SG transgene constitutively produced throughout myogenesis. At 5 weeks after the transplantation, the cell rates expressing fast-myosin heavy chain (MHC) exceeded slow-MHC in delta-SG(+) cells. Fifteen weeks after (corresponding to approximately 12 years in humans), fast MHC(+) cells nullified, but the delta-SG(+) and slow MHC(+) cell number remained unaltered. These skMbs fused with host cardiomyocytes via connexin-43 and intercalated disc, modestly improving the hemodynamics without arrhythmia, when engrafted skMbs were sparsely disseminated in autopsied myocardium. These results provide us evidence that disseminating delivery of slow-MHC(+) myoblasts is promising for repairing DCM heart using histocompatible skeletal myoblasts in future.


Asunto(s)
Cardiomiopatías/patología , Cardiomiopatías/cirugía , Músculo Esquelético/patología , Músculo Esquelético/trasplante , Mioblastos/patología , Mioblastos/trasplante , Miocitos Cardíacos/patología , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Cricetinae , Masculino , Regeneración/fisiología , Resultado del Tratamiento
11.
Int Heart J ; 47(2): 247-58, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16607052

RESUMEN

In hypertrophic cardiomyopathy (HCM) a hyperkinetic state is sometimes observed in spite of impaired systolic function in the hypertrophied myocardium. The aim of the present study was to determine the mechanism of this paradox. Seventeen patients with HCM and 10 normal subjects underwent cine magnetic resonance (MR) imaging to measure percent systolic wall thickening and percent fractional shortening. The ratio of systolic radial wall stress of the LV at the hypertrophied myocardium over that at the nonhypertrophied myocardium was evaluated to describe the focal advantageous condition for wall thickening. The ratio was 0.66 +/- 0.36 at the start of contraction and 0.78 +/- 0.31 at early-systole, indicating consistently smaller radial wall stress at the hypertrophied myocardium. Although the condition for contraction was favorable (a ratio less than 1.00), percent systolic wall thickening at the hypertrophied myocardium (23.0 +/- 11.8%) was smaller than that at the nonhypertrophied myocardium (70.5 +/- 32.3%). Smaller end-diastolic dimension (HCM group; 45.2 +/- 4.2 mm, reference group; 48.9 +/- 4.1 mm, P = 0.04) with a statistically identical value of systolic decrease in intraventricular dimension (HCM group; 19.7 +/- 3.9 mm, reference group; 18.9 +/- 3.2 mm, P = 0.60) yielded high percent fractional shortening in patients with HCM (43.5 +/- 7.6%). Although contractile impairment was proven at the hypertrophied region with low radial wall stress in the HCM group, the smaller end-diastolic dimension in this group resulted in high percent fractional shortening.


Asunto(s)
Cardiomiopatía Hipertrófica/fisiopatología , Imagen por Resonancia Cinemagnética , Contracción Miocárdica , Miocardio/patología , Función Ventricular Izquierda , Cardiomiopatía Hipertrófica/diagnóstico , Electrocardiografía , Humanos , Volumen Sistólico , Sístole
12.
Am J Pathol ; 168(2): 386-97, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16436654

RESUMEN

In UM-X7.1 hamster model of human dilated cardiomyopathy, heart failure progressively develops and causes 50% mortality by 30 weeks of age. Through ultrastructural analysis, we found that many cardiomyocytes of this model contain typical autophagic vacuoles including degraded mitochondria, glycogen granules, and myelin-like figures. In addition, ubiquitin, cathepsin D, and Rab7 were overexpressed as determined by immunoassays. Importantly, most cardiomyocytes with leaky plasma membranes were positive for cathepsin D, suggesting a direct link between autophagic degeneration and cell death. Meanwhile, cardiomyocyte apoptosis appeared insignificant. Granulocyte colony-stimulating factor (10 microg/kg/day), injected 5 days/week from 15 to 30 weeks of age, improved survival among 30-week-old hamsters (100% versus 53% in the untreated hamsters, P < 0.0001); ventricular function and remodeling, increased cardiomyocyte size, and reduced myocardial fibrosis followed by a dramatic reduction in the autophagic findings were also seen. Granulocyte colony-stimulating factor also down-regulated tumor necrosis factor-alpha and increased activities of Akt signal transducer and activator of transcription-3, and matrix metalloproteinases. However, there was no clear evidence of transdifferentiation from bone marrow cells into cardiomyocytes. In conclusion, autophagic death is important for cardiomyocyte loss in the cardiomyopathic hamster, and the beneficial effect of granulocyte colony-stimulating factor acts mainly via an anti-autophagic mechanism rather than anti-apo-ptosis or regeneration.


Asunto(s)
Apoptosis , Autofagia , Cardiomiopatía Dilatada/prevención & control , Fibrosis/prevención & control , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Miocitos Cardíacos/patología , Animales , Cardiomiopatía Dilatada/metabolismo , Catepsina D/metabolismo , Cricetinae , Fibrosis/metabolismo , Masculino , Mesocricetus , Mitocondrias/patología , Vaina de Mielina , Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Recombinantes , Factor de Transcripción STAT3/metabolismo , Tasa de Supervivencia , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitina/metabolismo , Vacuolas , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión a GTP rab7
13.
Life Sci ; 78(1): 22-9, 2005 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-16154157

RESUMEN

We examined the role of prostaglandin D(2) (PGD(2)) in the expression of vascular cell adhesion molecule-1 (VCAM)-1 following interleukin-1beta (IL-1) stimulation in human umbilical vein endothelial cells (HUVEC) transfected with lipocaline-type PGD(2) synthase (L-PGDS) genes. HUVEC were isolated from human umbilical vein and incubated with 20 U/ml IL-1 and various concentrations of authentic PGD(2). The isolated HUVEC were also transfected with L-PGDS genes by electroporation. The L-PGDS-transfected HUVEC were used to investigate the role of endogenous PGD(2) in IL-1-stimulated VCAM-1 biosynthesis. We also used an anti-PGD(2) antibody to examine whether an intracrine mechanism was involved in VCAM-1 production. PGD(2) and VCAM-1 levels were determined by radio- and cell surface enzyme-immunoassay, respectively. VCAM-1 mRNA was assessed by RT-PCR. IL-1-stimulated VCAM-1 expression by HUVEC was dose-dependently inhibited by authentic PGD(2). L-PGDS gene-transfected HUVEC produced more PGD(2) than HUVEC transfected with the reporter gene alone. IL-1 induced increases in VCAM-1 expression in HUVEC transfected with reporter genes alone. However, this effect was significantly attenuated in the case of IL-1 stimulation of HUVEC transfected with L-PGDS genes, and accompanied by an apparent suppression of VCAM-1 mRNA expression. Neutralization of extracellular PGD(2) by anti-PGD(2)-specific antibody influenced neither VCAM-1 mRNA expression nor VCAM-1 biosynthesis. In conclusion, HUVEC transfected with L-PGDS genes showed increased PGD(2) synthesis. This increase was associated with attenuation of both VCAM-1 expression and VCAM-1 mRNA expression. The results suggest that endogenous PGD(2) decreases VCAM-1 expression and VCAM-1 mRNA expression, probably through an intracrine mechanism.


Asunto(s)
Células Endoteliales/metabolismo , Prostaglandina D2/biosíntesis , Venas Umbilicales/metabolismo , Molécula 1 de Adhesión Celular Vascular/biosíntesis , Eicosanoides/análisis , Eicosanoides/biosíntesis , Femenino , Humanos , Interleucina-1/metabolismo , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/metabolismo , Lipocalinas , Prostaglandina D2/fisiología , ARN Mensajero/biosíntesis , Radioinmunoensayo , Transfección , Venas Umbilicales/citología
14.
Biochim Biophys Acta ; 1751(1): 73-81, 2005 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16054019

RESUMEN

The precise mechanism of the progression of advanced heart failure is unknown. We assessed a new scheme in two heart failure models: (I) congenital dilated cardiomyopathy (DCM) in TO-2 strain hamsters lacking delta-sarcoglycan (SG) gene and (II) administration of a high-dose of isoproterenol, as an acute heart failure in normal rats. In TO-2 hamsters, we followed the time course of the histological, physiological and metabolic the progressions of heart failure to the end stage. Dystrophin localization detected by immunostaining age-dependently to the myoplasm and the in situ sarcolemma fragility evaluated by Evans blue entry was increased in the same cardiomyocytes. Western blotting revealed a limited cleavage of the dystrophin protein at the rod domain, strongly suggesting a contribution of endogenous protease(s). We found a remarkable up-regulation of the amount of calpain-1 and -2, and no change of their counterpart, calpastatin. After supplementing TO-2 hearts with the normal delta-SG gene in vivo, these pathological alterations and the animals' survival improved. Furthermore, dystrophin but not delta-SG was disrupted by a high dose of isoproterenol, translocated from the sarcolemma to the myoplasm and fragmented. These results of heart failure, irrespective of the hereditary or acquired origin, indicate a vicious cycle formed by the increased sarcolemma permeability, preferential activation of calpain over calpastatin, and translocation and cleavage of dystrophin would commonly lead to advanced heart failure.


Asunto(s)
Cardiomiopatía Dilatada/fisiopatología , Distrofina/fisiología , Animales , Proteínas de Unión al Calcio/biosíntesis , Calpaína/metabolismo , Cardiomiopatía Dilatada/congénito , Cardiomiopatía Dilatada/terapia , Permeabilidad de la Membrana Celular , Cricetinae , Dependovirus/fisiología , Modelos Animales de Enfermedad , Activación Enzimática , Terapia Genética , Insuficiencia Cardíaca/inducido químicamente , Isoproterenol , Mesocricetus , Modelos Biológicos , Ratas , Sarcoglicanos/deficiencia , Sarcoglicanos/genética , Sarcolema/fisiología
15.
Pharmacol Ther ; 107(1): 31-43, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15963350

RESUMEN

The precise mechanism(s) of the progression of advanced heart failure (HF) should be determined to establish strategies for its treatment or prevention. Based on pathological, molecular, and physiological findings in 3 animal models and human cases, we propose a novel scheme that a vicious cycle formed by increased sarcolemma (SL) permeability, preferential activation of calpain over calpastatin, and translocation and cleavage of dystrophin (Dys) commonly lead to advanced HF. The aim of this article was to assess our recent paradigm that disruption of myocardial Dys is a final common pathway to advanced HF, irrespective of its hereditary or acquired origin, but not intended to provide a comprehensive overview of the various factors that may be involved in the course of HF in different clinical settings. In addition, each component of Dys-associated proteins (DAP) was heterogeneously degraded in vivo and in vitro, i.e. Dys and alpha-sarcoglycan (SG) were markedly destroyed using isolated calpain 2, while delta-SG was not degraded at all. The up-regulation of calpain 2 was confirmed through previously published data that remain insufficient for precise evaluation, supporting our new scheme that the activation of calpain(s) is involved in the steady process of Dys cleavage. In addition, somatic gene therapy is discussed as a potential option to ameliorate the physiological/metabolic indices and to improve the prognosis.


Asunto(s)
Calpaína/fisiología , Cardiomiopatía Dilatada/metabolismo , Modelos Animales de Enfermedad , Distrofina/fisiología , Terapia Genética/métodos , Insuficiencia Cardíaca , Sarcoglicanos/fisiología , Animales , Calpaína/efectos adversos , Calpaína/metabolismo , Cardiomiopatía Dilatada/fisiopatología , Distrofina/metabolismo , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/terapia , Humanos , Infarto del Miocardio/complicaciones , Sarcoglicanos/clasificación , Sarcoglicanos/metabolismo , Transducción Genética/métodos
16.
Br J Pharmacol ; 146(1): 49-59, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15937516

RESUMEN

The A-type voltage-dependent K(+) current (I(A)) has been identified in several types of smooth muscle cells including the pulmonary artery (PA), but little is known about the pharmacological and molecular characteristics of I(A) in human pulmonary arterial smooth muscle cells (hPASMCs). We investigated I(A) expressed in cultured PASMCs isolated from the human main pulmonary artery, using patch-clamp techniques, reverse transcriptase-polymerase chain reaction (RT-PCR), quantitative real-time RT-PCR and immunocytochemical studies. With high EGTA and ATP in the pipette, the outward currents were dominated by a transient K(+) current (I(A)), followed by a relatively small sustained outward current (I(K)). I(A) was inhibited by 4-aminopyridine (4-AP) concentration-dependently, and could be separated pharmacologically into two components by tetraethylammonium (TEA) sensitivity. A component was sensitive to TEA, and the second component was insensitive to TEA. I(A) was inhibited by blood depressing substrate (BDS)-II, a specific blocker of K(V)3.4 subunit, and phrixotoxin-II, a specific blocker of K(V)4.2 and 4.3. Flecainide inhibited I(A) concentration-dependently, but it inhibited it preferentially in the presence of TEA (TEA-insensitive I(A)). Systematic screening of expression of K(V) genes using RT-PCR showed the definite presence of transcripts of the I(A)-encoding genes for K(V)3.4, K(V)4.1, K(V)4.2 and K(V)4.3 as well as the I(K)-encoding genes for K(V)1.1, K(V)1.5 and K(V)2.1. The real-time RT-PCR analysis showed that the relative abundance of the encoding genes of I(A) alpha-subunit and K(V) channel-interacting proteins (KChIPs) was K(V)4.2 > K(V)3.4 > K(V)4.3 (long) > K(V)4.1, and KChIP3 >> KChIP2, respectively. The presence of K(V)3.4, K(V)4.2 and K(V)4.3 proteins was also demonstrated by immunocytochemical studies, and confirmed by immunohistochemical staining using intact human PA sections. These results suggest that I(A) in cultured hPASMCs consists of two kinetically and pharmacologically distinct components, probably K(V)3.4 and K(V)4 channels.


Asunto(s)
Potenciales de la Membrana/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Canales de Potasio con Entrada de Voltaje/efectos de los fármacos , Canales de Potasio con Entrada de Voltaje/genética , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Células Cultivadas , Regulación de la Expresión Génica , Inmunohistoquímica , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/fisiología , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/fisiología , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/fisiología , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Cardiovasc Res ; 65(2): 356-65, 2005 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-15639474

RESUMEN

OBJECTIVES: Genetic depletion of dystrophin-related protein (DRP) complex causes cardiomyopathy in animals and humans. We found in a previous study that some types of DRP were degraded and that calpain content was increased in rats with non-genetically induced heart failure. The present study was aimed at examining the effects of an angiotensin-I-converting enzyme inhibitor (ACEI) trandolapril (Tra) or an angiotensin II type 1 receptor blocker (ARB) candesartan (Can), both of which are known to improve the pathophysiology of chronic heart failure (CHF) on degradation of DRP in failing hearts. METHODS: Coronary artery-ligated (CAL) and sham-operated rats (Sham rats) were treated orally with 3 mg/kg/day trandolapril (Tra) or 1 mg/kg/day candesartan (Can) from the 2nd to 8th week after surgery. RESULTS: Hemodynamic parameters of CAL rats at the 8th week after CAL (8w-CAL) indicated heart failure. alpha-Sarcoglycan (SG) and dystrophin in the surviving left ventricle (surviving LV) of 8w-CAL rats decreased, whereas beta-, gamma-, and delta-SGs remained unchanged. Calcium-activated neutral proteases mu-calpain and m-calpain increased in the surviving LV at the 8th week of postmyocardial infarction. Proteolytic activity in the presence of 5 mM Ca2+ markedly increased at the 2nd and 8th weeks, whereas 50 microM Ca2+ slightly but significantly increased proteolysis of casein. Tra or Can treatment improved the hemodynamic parameters, attenuated changes in alpha-SG and dystrophin, and reversed both calpain contents and activities of the failing heart back to sham levels. CONCLUSION: These results suggest that attenuation in calpain-induced degradation of DRP complex is a possible mechanism for the Tra- or Can-mediated improvement of the pathogenesis of CHF following myocardial infarction.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Calpaína/metabolismo , Insuficiencia Cardíaca/metabolismo , Miocardio/metabolismo , Receptor de Angiotensina Tipo 1/efectos de los fármacos , Sarcoglicanos/metabolismo , Animales , Bencimidazoles/farmacología , Compuestos de Bifenilo , Western Blotting/métodos , Calpaína/análisis , Citosol/química , Citosol/metabolismo , Hemodinámica/efectos de los fármacos , Indoles/farmacología , Masculino , Modelos Animales , Miocardio/química , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sarcoglicanos/análisis , Tetrazoles/farmacología
18.
Eur J Pharmacol ; 505(1-3): 67-74, 2004 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-15556138

RESUMEN

Endothelin-1 is known to be implicated in the pathogenesis of hepatobiliary diseases such as cirrhosis, especially in portal hypertension. This study aimed to investigate the effects of ursodeoxycholic acid on endothelin-1 production in human endothelial cells. The effects of ursodeoxycholic acid and its conjugates (tauroursodeoxycholic and glycoursodeoxycholic acids) on endothelin-1 production as well as nitric oxide (NO) in human umbilical vein endothelial cells (HUVECs) were examined. The production of endothelin-1 and nitric oxide in culture medium was measured using enzyme-linked immunosorbent assay (ELISA) and the Griess method, respectively. Endothelin-1 and endothelial nitric oxide synthase (eNOS) mRNA expression were investigated by real-time quantitative reverse transcriptase/polymerase chain reaction (RT-PCR). Ursodeoxycholic acid (30-1000 microM) inhibited endothelin-1 production in a concentration-dependent manner, and ursodeoxycholic acid at concentrations higher than 300 microM increased nitric oxide production in culture medium. The conjugates of ursodeoxycholic acid also increased nitric oxide production and decreased endothelin-1 production, which was less effective than ursodeoxycholic acid. N-nitro-L-arginine-mythel-ester (L-NAME), a nitric oxide synthase (NOS) inhibitor, suppressed the ursodeoxycholic acid-induced nitric oxide production, but it did not antagonize the inhibitory effects of ursodeoxycholic acid on endothelin-1 production. Ursodeoxycholic acid also induced a concentration-dependent decrease in endothelin-1 mRNA expression without significant changes in eNOS mRNA expression. These results provide novel evidence that ursodeoxycholic acid inhibits endothelin-1 production in human endothelial cells, but nitric oxide is not responsible for the inhibitory effect of ursodeoxycholic acid on endothelin-1. Thus, ursodeoxycholic acid therapy may prevent the development of several pathogenesis such as portal hypertension observed in patients with cirrhosis due to the improvement of endothelial function.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Endotelina-1/biosíntesis , Ácido Ursodesoxicólico/farmacología , Línea Celular , Relación Dosis-Respuesta a Droga , Células Endoteliales/metabolismo , Endotelina-1/genética , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática , Expresión Génica/efectos de los fármacos , Humanos , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo III , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Proc Natl Acad Sci U S A ; 101(19): 7381-5, 2004 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-15128945

RESUMEN

Advanced heart failure (HF) is the leading cause of death in developed countries. The mechanism underlying the progression of cardiac dysfunction needs to be clarified to establish approaches to prevention or treatment. Here, using TO-2 hamsters with hereditary dilated cardiomyopathy, we show age-dependent cleavage and translocation of myocardial dystrophin (Dys) from the sarcolemma (SL) to the myoplasm, increased SL permeability in situ, and a close relationship between the loss of Dys and hemodynamic indices. In addition, we observed a surprising correlation between the amount of Dys and the survival rate. Dys disruption is not an epiphenomenon but directly precedes progression to advanced HF, because long-lasting transfer of the missing delta-SG gene to degrading cardiomyocytes in vivo with biologically nontoxic recombinant adenoassociated virus (rAAV) vector ameliorated all of the pathological features and changed the disease prognosis. Furthermore, acute HF after isoproterenol toxicity and chronic HF after coronary ligation in rats both time-dependently cause Dys disruption in the degrading myocardium. Dys cleavage was also detected in human hearts from patients with dilated cardiomyopathy of unidentified etiology, supporting a scheme consisting of SL instability, Dys cleavage, and translocation of Dys from the SL to the myoplasm, irrespective of an acute or chronic disease course and a hereditary or acquired origin. Hereditary HF may be curable with gene therapy, once the responsible gene is identified and precisely corrected.


Asunto(s)
Distrofina/metabolismo , Miocardio/metabolismo , Animales , Hemodinámica , Humanos , Hidrólisis , Masculino , Transporte de Proteínas , Ratas
20.
Biochem Biophys Res Commun ; 319(1): 50-7, 2004 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-15158441

RESUMEN

Dicer plays an important role in the course of RNA interference (RNAi), i.e., it digests long double-stranded RNAs into 21-25 nucleotide small-interfering RNA (siRNA) duplexes functioning as sequence-specific RNAi mediators. In this study, we investigated the expression levels of Dicer and eIF2C1 approximately 4, which, like Dicer, appear to participate in mammalian RNAi, in various mouse tissues. Results indicate that the levels of eIF2C1 approximately 4 as well as Dicer are lower in skeletal muscle and heart than in other tissues. To see if RNAi could occur under such a condition with low levels of expression of Dicer and eIF2C1 approximately 4, we examined RNAi activity in mouse skeletal muscle fibers. The results indicate that RNAi can be induced by synthetic siRNA duplexes in muscle fibers. We further examined RNAi activity during myogenic differentiation of mouse C2C12 cells. The data indicate that although the expression levels of Dicer and eIF2C1 approximately 4 decrease during the differentiation, RNAi can be induced in the cells. Altogether, the data presented here suggest that muscle cells retain the ability to induce RNAi, although Dicer and eIF2C1 approximately 4 appear to be barely expressed in them.


Asunto(s)
Endorribonucleasas/biosíntesis , Factores Eucarióticos de Iniciación/biosíntesis , Músculos/metabolismo , ARN Helicasas/biosíntesis , Interferencia de ARN , Animales , Proteínas Argonautas , Bromodesoxiuridina/farmacología , Ciclo Celular , Diferenciación Celular , Línea Celular , Colorantes/farmacología , ARN Helicasas DEAD-box , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Genes Reporteros , Luciferasas/metabolismo , Ratones , Músculo Esquelético/metabolismo , Oligonucleótidos/química , Biosíntesis de Proteínas , ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleasa III , Factores de Tiempo , Distribución Tisular , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...