Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Banco de datos
Tipo del documento
Publication year range
1.
Am J Physiol Heart Circ Physiol ; 309(5): H867-79, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26071548

RESUMEN

Previous studies have demonstrated the protective signaling of hypoxia-inducible factor (HIF)-1 α against ischemia-reperfusion (I/R) injury in the heart. In the present study, we provide further evidence for a cardioprotective mechanism by HIF-1α against I/R injury exerted via the mitochondrial protein frataxin, which regulates mitochondrial Fe-S cluster formation. Disruption of frataxin has been found to induce mitochondrial iron overload and subsequent ROS production. We observed that frataxin expression was elevated in mice hearts subjected to I/R injury, and this response was blunted in cardiomyocyte-specific HIF-1α knockout (KO) mice. Furthermore, these HIF-1α KO mice sustained extensive cardiac damage from I/R injury compared with control mice. Similarly, reduction of HIF-1α by RNA inhibition resulted in an attenuation of frataxin expression in response to hypoxia in H9C2 cardiomyocytes. Therefore, we postulated that HIF-1α transcriptionally regulates frataxin expression in response to hypoxia and offers a cardioprotective mechanism against ischemic injury. Our promoter activity and chromatin immunoprecipitation assays confirmed the presence of a functional hypoxia response element in the frataxin promoter. Our data also suggest that increased frataxin mitigated mitochondrial iron overload and subsequent ROS production, thus preserving mitochondrial membrane integrity and viability of cardiomyocytes. We postulate that frataxin may exert its beneficial effects by acting as an iron storage protein under hypoxia and subsequently facilitates the maintenance of mitochondrial membrane potential and promotes cell survival. The findings from our study revealed that HIF-1α-frataxin signaling promotes a protective mechanism against hypoxic/ischemic stress.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteínas de Unión a Hierro/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Animales , Células Cultivadas , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/crecimiento & desarrollo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Proteínas de Unión a Hierro/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Sarcómeros/metabolismo , Sarcómeros/ultraestructura , Transducción de Señal , Frataxina
2.
Am J Physiol Heart Circ Physiol ; 309(5): H844-59, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26209053

RESUMEN

Doxorubicin (DOX) is a highly effective anti-neoplastic agent; however, its cumulative dosing schedules are clinically limited by the development of cardiotoxicity. Previous studies have attributed the cause of DOX-mediated cardiotoxicity to mitochondrial iron accumulation and the ensuing reactive oxygen species (ROS) formation. The present study investigates the role of frataxin (FXN), a mitochondrial iron-sulfur biogenesis protein, and its role in development of DOX-mediated mitochondrial dysfunction. Athymic mice treated with DOX (5 mg/kg, 1 dose/wk with treatments, followed by 2-wk recovery) displayed left ventricular hypertrophy, as observed by impaired cardiac hemodynamic performance parameters. Furthermore, we also observed significant reduction in FXN expression in DOX-treated animals and H9C2 cardiomyoblast cell lines, resulting in increased mitochondrial iron accumulation and the ensuing ROS formation. This observation was paralleled in DOX-treated H9C2 cells by a significant reduction in the mitochondrial bioenergetics, as observed by the reduction of myocardial energy regulation. Surprisingly, similar results were observed in our FXN knockdown stable cell lines constructed by lentiviral technology using short hairpin RNA. To better understand the cardioprotective role of FXN against DOX, we constructed FXN overexpressing cardiomyoblasts, which displayed cardioprotection against mitochondrial iron accumulation, ROS formation, and reduction of mitochondrial bioenergetics. Lastly, our FXN overexpressing cardiomyoblasts were protected from DOX-mediated cardiac hypertrophy. Together, our findings reveal novel insights into the development of DOX-mediated cardiomyopathy.


Asunto(s)
Cardiomegalia/metabolismo , Doxorrubicina/efectos adversos , Proteínas de Unión a Hierro/metabolismo , Animales , Cardiomegalia/etiología , Cardiotoxicidad , Línea Celular , Células Cultivadas , Hierro/metabolismo , Proteínas de Unión a Hierro/genética , Ratones , Mitocondrias Cardíacas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Frataxina
3.
Pharm Res ; 32(3): 852-62, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25158648

RESUMEN

PURPOSE: Cardiotoxicity associated with the use of doxorubicin (DOX), and other chemotherapeutics, limits their clinical potential. This study determined the pharmacokinetics and antitumor and cardioprotective activity of free and liposome encapsulated phenyl-2-aminoethyl-selenide (PAESe). METHODS: The pharmacokinetics of free PAESe and PAESe encapsulated in liposomes (SSL-PAESe) were determined in rats using liquid chromatography tandem mass-spectrometry. The antitumor and cardioprotective effects were determined in a mouse xenograft model of human prostate (PC-3) cancer and cardiomyocytes (H9C2). RESULTS: The encapsulation of PAESe in liposomes increased the circulation half-life and area under the drug concentration time profile, and decreased total systemic clearance significantly compared to free PAESe. Free- and SSL-PAESe improved survival, decreased weight-loss and prevented cardiac hypertrophy significantly in tumor bearing and healthy mice following treatment with DOX at 5 and 12.5 mg/kg. In vitro studies revealed PAESe treatment altered formation of reactive oxygen species (ROS), cardiac hypertrophy and gene expression, i.e., atrial natriuretic peptide and myosin heavy chain complex beta, in H9C2 cells. CONCLUSIONS: Treatment with free and SSL-PAESe exhibited antitumor activity in a prostate xenograft model and mitigated DOX-mediated cardiotoxicity.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Antioxidantes/administración & dosificación , Antioxidantes/farmacocinética , Cardiomegalia/prevención & control , Etilaminas/administración & dosificación , Etilaminas/farmacocinética , Miocitos Cardíacos/efectos de los fármacos , Compuestos de Organoselenio/administración & dosificación , Compuestos de Organoselenio/farmacocinética , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Antineoplásicos/química , Antioxidantes/química , Área Bajo la Curva , Cardiomegalia/inducido químicamente , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patología , Línea Celular Tumoral , Química Farmacéutica , Cromatografía Liquida , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Doxorrubicina , Etilaminas/química , Regulación de la Expresión Génica/efectos de los fármacos , Semivida , Humanos , Inyecciones Intravenosas , Liposomas , Masculino , Espectrometría de Masas , Tasa de Depuración Metabólica , Ratones Desnudos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Compuestos de Organoselenio/química , Estrés Oxidativo/efectos de los fármacos , Neoplasias de la Próstata/patología , Ratas Endogámicas F344 , Especies Reactivas de Oxígeno/metabolismo , Tecnología Farmacéutica/métodos , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Front Pharmacol ; 11: 574656, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33912028

RESUMEN

Doxorubicin (DOX) is an anthracycline cancer chemotherapeutic that exhibits cumulative dose-limiting cardiotoxicity and limits its clinical utility. DOX treatment results in the development of morbid cardiac hypertrophy that progresses to congestive heart failure and death. Recent evidence suggests that during the development of DOX mediated cardiac hypertrophy, mitochondrial energetics are severely compromised, thus priming the cardiomyocyte for failure. To mitigate cumulative dose (5 mg/kg, QIW x 4 weeks with 2 weeks recovery) dependent DOX, mediated cardiac hypertrophy, we applied an orally active selenium based compound termed phenylaminoethyl selenides (PAESe) (QIW 10 mg/kg x 5) to our animal model and observed that PAESe attenuates DOX-mediated cardiac hypertrophy in athymic mice, as observed by MRI analysis. Mechanistically, we demonstrated that DOX impedes the stability of the iron-sulfur cluster biogenesis protein Frataxin (FXN) (0.5 fold), resulting in enhanced mitochondrial free iron accumulation (2.5 fold) and reduced aconitase activity (0.4 fold). Our findings further indicate that PAESe prevented the reduction of FXN levels and the ensuing elevation of mitochondrial free iron levels. PAESe has been shown to have anti-oxidative properties in part, by regeneration of glutathione levels. Therefore, we observed that PAESe can mitigate DOX mediated cardiac hypertrophy by enhancing glutathione activity (0.4 fold) and inhibiting ROS formation (1.8 fold). Lastly, we observed that DOX significantly reduced cellular respiration (basal (5%) and uncoupled (10%)) in H9C2 cardiomyoblasts and that PAESe protects against the DOX-mediated attenuation of cellular respiration. In conclusion, the current study determined the protective mechanism of PAESe against DOX mediated myocardial damage and that FXN is implicitly involved in DOX-mediated cardiotoxicity.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda