Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Toxicol Appl Pharmacol ; 453: 116210, 2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-36028075

RESUMEN

Doxorubicin (DOX) is an effective anthracycline used in chemotherapeutic regimens for a variety of haematological and solid tumors. However, its utility remains limited by its well-described, but poorly understood cardiotoxicity. Despite numerous studies describing various forms of regulated cell death and their involvement in DOX-mediated cardiotoxicity, the predominate form of cell death remains unclear. Part of this inconsistency lies in a lack of standardization of in vivo and in vitro model design. To this end, the objective of this study was to characterize acute low- and high-dose DOX exposure on cardiac structure and function in C57BL/6 N mice, and evaluate regulated cell death pathways and autophagy both in vivo and in cardiomyocyte culture models. Acute low-dose DOX had no significant impact on cardiac structure or function; however, acute high-dose DOX elicited substantial cardiac necrosis resulting in diminished cardiac mass and volume, with a corresponding reduced cardiac output, and without impacting ejection fraction or fibrosis. Low-dose DOX consistently activated caspase-signaling with evidence of mitochondrial permeability transition. However, acute high-dose DOX had only modest impact on common necrotic signaling pathways, but instead led to an inhibition in autophagic flux. Intriguingly, when autophagy was inhibited in cultured cardiomyoblasts, DOX-induced necrosis was enhanced. Collectively, these observations implicate inhibition of autophagy flux as an important component of the acute necrotic response to DOX, but also suggest that acute high-dose DOX exposure does not recapitulate the disease phenotype observed in human cardiotoxicity.


Asunto(s)
Cardiotoxicidad , Doxorrubicina , Animales , Antibióticos Antineoplásicos/toxicidad , Apoptosis , Autofagia , Cardiotoxicidad/metabolismo , Muerte Celular , Doxorrubicina/farmacología , Humanos , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos , Necrosis
2.
Biochem Cell Biol ; 93(5): 452-65, 2015 10.
Artículo en Inglés | MEDLINE | ID: mdl-26151290

RESUMEN

The prevalence of type 2 diabetes (T2D) has increased dramatically over the past two decades, not only among adults but also among adolescents. T2D is a systemic disorder affecting every organ system and is especially damaging to the cardiovascular system, predisposing individuals to severe cardiac and vascular complications. The precise mechanisms that cause T2D are an area of active research. Most current theories suggest that the process begins with peripheral insulin resistance that precedes failure of the pancreatic ß-cells to secrete sufficient insulin to maintain normoglycemia. A growing body of literature has highlighted multiple aspects of mitochondrial function, including oxidative phosphorylation, lipid homeostasis, and mitochondrial quality control in the regulation of peripheral insulin sensitivity. Whether the cellular mechanisms of insulin resistance in adults are comparable to that in adolescents remains unclear. This review will summarize both clinical and basic studies that shed light on how alterations in skeletal muscle mitochondrial function contribute to whole body insulin resistance and will discuss the evidence supporting high-intensity exercise training as a therapy to circumvent skeletal muscle mitochondrial dysfunction to restore insulin sensitivity in both adults and adolescents.


Asunto(s)
Diabetes Mellitus Tipo 2/prevención & control , Mitocondrias Musculares/metabolismo , Músculo Esquelético/metabolismo , Adolescente , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Resistencia a la Insulina , Músculo Esquelético/patología
3.
J Pediatr Gastroenterol Nutr ; 61(5): 583-90, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26505960

RESUMEN

OBJECTIVES: The aim of the present study was to determine the in vitro effect(s) of a bovine-based human breast milk fortifier (HMF) on human intestinal cells. HMF increases the expression of BCL2/adenovirus E1B 19 kDa protein-interacting protein (Bnip3) and cell death; the prostaglandin analogue misoprostol will rescue this effect. METHODS: Cultured intestinal cells were exposed to in vitro-digested human breast milk (BM) ±â€ŠHMF. Intracellular oxidation, cell damage/cell death, and BNIP3 expression were measured after exposure. RESULTS: In vitro-digested BM + HMF significantly increased intracellular oxidation, cell damage, and cell death in enterocyte cell cultures compared with either saline or BM controls, an effect that was rescued by the prostaglandin analogue, misoprostol. Bnip3 transcript and Bnip3 protein levels were significantly increased in vitro after treatment with BM + HMF. We also provide evidence that transfection of enterocytes with Bnip3 increases cell death, an effect that is rescued by a nonfunctional Bnip3 splice variant. CONCLUSIONS: Our data support the hypothesis that HMF increases intestinal Bnip3 in vitro, and that the gene product triggers cell death. We suggest that misoprostol is a promising therapy, which may reduce intestinal cell death.


Asunto(s)
Muerte Celular , Suplementos Dietéticos/efectos adversos , Alimentos Fortificados/efectos adversos , Fórmulas Infantiles/química , Intestinos/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Leche Humana , Proteínas Proto-Oncogénicas/metabolismo , Animales , Bovinos , Línea Celular , Dieta , Enterocolitis Necrotizante/etiología , Enterocolitis Necrotizante/prevención & control , Enterocitos/metabolismo , Femenino , Humanos , Técnicas In Vitro , Lactante , Recien Nacido Prematuro , Mucosa Intestinal/metabolismo , Intestinos/citología , Proteínas Mitocondriales/metabolismo , Transfección
4.
Curr Hypertens Rep ; 14(6): 540-7, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23001875

RESUMEN

Despite the progress that has been made over the past two decades in cardiovascular research, heart failure remains a major cause of morbidity and mortality worldwide. Insight into the cellular and molecular mechanisms that underlie the heart failure in individuals with ischemic heart disease have identified defects in cellular processes that govern autophagy, apoptosis and necrosis as a prevailing underlying cause. Indeed, programmed cell death of cardiac cells by apoptosis or necrosis is believed to involve the intrinsic mitochondrial pathway and/or extrinsic death receptor pathway by certain Bcl-2 family members as well as components of the TNFα signaling pathway. In this review, we discuss recent advances in the molecular signaling factors that govern cardiac cell fate under normal and disease conditions.


Asunto(s)
Apoptosis/fisiología , Autofagia/fisiología , Cardiopatías/metabolismo , Necrosis/metabolismo , Humanos , Mitocondrias/metabolismo , Miocitos Cardíacos/metabolismo , Transducción de Señal
5.
Pediatr Cardiol ; 32(3): 263-6, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21293853

RESUMEN

A delicate balance exists between cell growth and cell death. In the context of the adult myocardium, inappropriate or inordinate cell loss through an apoptotic process may profoundly influence cardiac structure, function, or both given the limited and meager ability of the heart for repair after injury. Earlier work by the authors' laboratory identified a close relation between cell cycle factor E2F-1 and hypoxia-inducible factor Bnip3 as the key regulator of apoptosis and autophagy in ventricular myocytes. Epigenetic changes by histone-modifying proteins, namely, histone deacetylases (HDACs) influence cell survival by altering the activity of histone core proteins, transcription factors, or both. This report highlights the intricate nature between the cellular factors E2F-1 and nuclear factor-κB (NF-κB) and the epigenetic regulation of Bnip3 gene transcription by HDAC1 for cell survival of ventricular myocytes.


Asunto(s)
Muerte Celular/genética , Epigénesis Genética , Miocitos Cardíacos/metabolismo , Factores de Transcripción E2F/metabolismo , Histona Desacetilasa 1/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas/metabolismo
7.
Cell Death Discov ; 4: 37, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30275982

RESUMEN

The cellular response to hypoxia involves the activation of a conserved pathway for gene expression regulated by the transcription factor complex called hypoxia-inducible factor (HIF). This pathway has been implicated in both the adaptive response to hypoxia and in several hypoxic-ischemic-related pathologies. Perinatal hypoxic injury, often associated with prematurity, leads to multi-organ dysfunction resulting in significant morbidity and mortality. Using a rodent model of neonatal hypoxia and several representative cell lines, we observed HIF1α activation and down-stream induction of the cell death gene Bnip3 in brain, large intestine, and heart which was mitigated by administration of the prostaglandin E1 analog misoprostol. Mechanistically, we determined that misoprostol inhibits full-length Bnip3 (Bnip3-FL) expression through PKA-mediated NF-κB (P65) nuclear retention, and the induction of pro-survival splice variants. We observed that the dominant small pro-survival variant of Bnip3 in mouse cells lacks the third exon (Bnip3ΔExon3), whereas human cells produce a pro-survival BNIP3 variant lacking exon 2 (BNIP3ΔExon2). In addition, these small Bnip3 splice variants prevent mitochondrial dysfunction, permeability transition, and necrosis triggered by Bnip3-FL by blocking calcium transfer from the sarco/endoplasmic reticulum to the mitochondria. Furthermore, misoprostol and Bnip3ΔExon3 promote nuclear calcium accumulation, resulting in HDAC5 nuclear export, NFAT activation, and adaptive changes in cell morphology and gene expression. Collectively, our data suggests that misoprostol can mitigate the potential damaging effects of hypoxia on multiple cell types by activating adaptive cell survival pathways through Bnip3 repression and alternative splicing.

8.
Cell Death Differ ; 25(10): 1732-1748, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29511336

RESUMEN

Myocardin is a transcriptional co-activator required for cardiovascular development, but also promotes cardiomyocyte survival through an unclear molecular mechanism. Mitochondrial permeability transition is implicated in necrosis, while pore closure is required for mitochondrial maturation during cardiac development. We show that loss of myocardin function leads to subendocardial necrosis at E9.5, concurrent with elevated expression of the death gene Nix. Mechanistically, we demonstrate that myocardin knockdown reduces microRNA-133a levels to allow Nix accumulation, leading to mitochondrial permeability transition, reduced mitochondrial respiration, and necrosis. Myocardin knockdown elicits calcium release from the endo/sarcoplasmic reticulum with mitochondrial calcium accumulation, while restoration of microRNA-133a function, or knockdown of Nix rescues calcium perturbations. We observed reduced myocardin and elevated Nix expression within the infarct border-zone following coronary ligation. These findings identify a myocardin-regulated pathway that maintains calcium homeostasis and mitochondrial function during development, and is attenuated during ischemic heart disease. Given the diverse role of Nix and microRNA-133a, these findings may have broader implications to metabolic disease and cancer.


Asunto(s)
Calcio/metabolismo , Mitocondrias/metabolismo , Proteínas Nucleares/metabolismo , Transactivadores/metabolismo , Animales , Células Cultivadas , Doxorrubicina/farmacología , Expresión Génica/efectos de los fármacos , Corazón/efectos de los fármacos , Isoproterenol/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Permeabilidad/efectos de los fármacos , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Retículo Sarcoplasmático/metabolismo , Transactivadores/antagonistas & inhibidores , Transactivadores/genética
9.
Exp Clin Cardiol ; 16(4): 102-8, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22131851

RESUMEN

Cardiac disease is a global epidemic that is on the rise, despite the recent advances in cardiovascular research. Once the myocardium is injured, it has a limited capacity to activate reparative mechanisms to restore proper cardiac function, leading to the development of systemic heart failure. Autophagy, under certain conditions, may result in cell death, further emphasizing the controversial issues regarding the autophagic process as an adaptive or maladaptive biological response. Although significant progress in understanding the signalling mechanisms of cell death in myocytes has been made, the role of apoptotic cell death and programmed necrosis during heart failure is not completely understood. Insight to how myocytes determine whether to activate apoptotic or programmed necrosis signalling machinery remains under current investigation because it is a major problem for both scientists and clinicians in treating heart failure patients. Herein, the different modes of cell death implicated in heart failure are highlighted, as well as the role of B-cell lymphoma-2 family members and how mitochondria act as central organelles in directing such cell death mechanisms.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA