Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Intervalo de año de publicación
2.
Expert Rev Cardiovasc Ther ; 14(9): 1007-19, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27308848

RESUMEN

INTRODUCTION: Myocardial infarction (MI) is the leading cause of death. When MI is not lethal, heart failure (HF) is a major consequence with high prevalence and poor prognosis. The targeting of autophagy represents a potentially therapeutic approach for the treatment of both pathologies. AREAS COVERED: PubMed searches were performed to discuss the current state of the art regarding the role of autophagy in MI and HF. We review available and potential approaches to modulate autophagy from a pharmacological and genetic perspective. We also discuss the targeting of autophagy in myocardial regeneration. Expert commentary: The targeting of autophagy has potential for the treatment of MI and HF. Autophagy is a process that takes place in virtually all cells of the body and thus, in order to evaluate this therapeutic approach in clinical trials, strategies that specifically target this process in the myocardium is required to avoid unwanted effects in other organs.


Asunto(s)
Autofagia , Insuficiencia Cardíaca/terapia , Infarto del Miocardio/terapia , Animales , Humanos , Miocardio/patología
3.
Pharmacol Res ; 103: 318-27, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26607864

RESUMEN

The alpha2-adrenergic receptor agonist Dexmedetomidine (Dex) is a sedative medication used by anesthesiologists. Dex protects the heart against ischemia-reperfusion (IR) and can also act as a preconditioning mimetic. The mechanisms involved in Dex-dependent cardiac preconditioning, and whether this action occurs directly or indirectly on cardiomyocytes, still remain unclear. The endothelial nitric oxide synthase (eNOS)/nitric oxide (NO) signaling pathway and endothelial cells are known to play key roles in cardioprotection against IR injury. Therefore, the aims of this work were to evaluate whether the eNOS/NO pathway mediates the pharmacological cardiac effect of Dex, and whether endothelial cells are required in this cardioprotective action. Isolated adult rat hearts were treated with Dex (10nM) for 25min and the dimerization of eNOS and production of NO were measured. Hearts were then subjected to global IR (30/120min) and the role of the eNOS/NO pathway was evaluated. Dex promoted the activation of eNOS and production of NO. Dex reduced the infarct size and improved the left ventricle function recovery, but this effect was reversed when Dex was co-administered with inhibitors of the eNOS/NO/PKG pathway. In addition, Dex was unable to reduce cell death in isolated adult rat cardiomyocytes subjected to simulated IR. Cardiomyocyte death was attenuated by co-culturing them with endothelial cells pre-treated with Dex. In summary, our results show that Dex triggers cardiac protection by activating the eNOS/NO signaling pathway. This pharmacological effect of Dex requires its interaction with the endothelium.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Cardiotónicos/farmacología , Dexmedetomidina/farmacología , Daño por Reperfusión Miocárdica/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/metabolismo , Agonistas de Receptores Adrenérgicos alfa 2/uso terapéutico , Animales , Cardiotónicos/uso terapéutico , Células Cultivadas , Técnicas de Cocultivo , Dexmedetomidina/uso terapéutico , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Corazón/efectos de los fármacos , Corazón/fisiopatología , Humanos , Masculino , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Ratas Sprague-Dawley
4.
J Mol Cell Cardiol ; 75: 40-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24997440

RESUMEN

In cardiomyocytes, Ca(2+) plays a central role in governing both contraction and signaling events that regulate gene expression. Current evidence indicates that discrimination between these two critical functions is achieved by segregating Ca(2+) within subcellular microdomains: transcription is regulated by Ca(2+) release within nuclear microdomains, and excitation-contraction coupling is regulated by cytosolic Ca(2+). Accordingly, a variety of agonists that control cardiomyocyte gene expression, such as endothelin-1, angiotensin-II or insulin-like growth factor-1, share the feature of triggering nuclear Ca(2+) signals. However, signaling pathways coupling surface receptor activation to nuclear Ca(2+) release, and the phenotypic responses to such signals, differ between agonists. According to earlier hypotheses, the selective control of nuclear Ca(2+) signals by activation of plasma membrane receptors relies on the strategic localization of inositol trisphosphate receptors at the nuclear envelope. There, they mediate Ca(2+) release from perinuclear Ca(2+) stores upon binding of inositol trisphosphate generated in the cytosol, which diffuses into the nucleus. More recently, identification of such receptors at nuclear membranes or perinuclear sarcolemmal invaginations has uncovered novel mechanisms whereby agonists control nuclear Ca(2+) release. In this review, we discuss mechanisms for the selective control of nuclear Ca(2+) signals with special focus on emerging models of agonist receptor activation.


Asunto(s)
Señalización del Calcio , Núcleo Celular/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Canales de Calcio/metabolismo , Citosol/metabolismo , Humanos , Modelos Biológicos
5.
Toxicol Appl Pharmacol ; 279(1): 53-62, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24844443

RESUMEN

RATIONALE: Dihydropyridines are widely used for the treatment of several cardiac diseases due to their blocking activity on L-type Ca(2+) channels and their renowned antioxidant properties. METHODS: We synthesized six novel dihydropyridine molecules and performed docking studies on the binding site of the L-type Ca(2+) channel. We used biochemical techniques on isolated adult rat cardiomyocytes to assess the efficacy of these molecules on their Ca(2+) channel-blocking activity and antioxidant properties. The Ca(2+) channel-blocking activity was evaluated by confocal microscopy on fluo-3AM loaded cardiomyocytes, as well as using patch clamp experiments. Antioxidant properties were evaluated by flow cytometry using the ROS sensitive dye 1,2,3 DHR. RESULTS: Our docking studies show that a novel compound with 3-OH substitution inserts into the active binding site of the L-type Ca(2+) channel previously described for nitrendipine. In biochemical assays, the novel meta-OH group in the aryl in C4 showed a high blocking effect on L-type Ca(2+) channel as opposed to para-substituted compounds. In the tests we performed, none of the molecules showed antioxidant properties. CONCLUSIONS: Only substitutions in C2, C3 and C5 of the aryl ring render dihydropyridine compounds with the capacity of blocking LTCC. Based on our docking studies, we postulate that the antioxidant activity requires a larger group than the meta-OH substitution in C2, C3 or C5 of the dihydropyridine ring.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/efectos de los fármacos , Dihidropiridinas/farmacología , Miocitos Cardíacos/efectos de los fármacos , Animales , Sitios de Unión , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/química , Cardiotónicos/farmacología , Separación Celular , Supervivencia Celular/efectos de los fármacos , Dihidropiridinas/química , Frecuencia Cardíaca/efectos de los fármacos , Hidroxilación , Masculino , Modelos Moleculares , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Relación Estructura-Actividad
6.
Trends Endocrinol Metab ; 25(3): 128-37, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24380833

RESUMEN

Insulin-like growth factor 1 (IGF-1) signaling regulates contractility, metabolism, hypertrophy, autophagy, senescence, and apoptosis in the heart. IGF-1 deficiency is associated with an increased risk of cardiovascular disease, whereas cardiac activation of IGF-1 receptor (IGF-1R) protects from the detrimental effects of a high-fat diet and myocardial infarction. IGF-1R activates multiple pathways through its intrinsic tyrosine kinase activity and through coupling to heterotrimeric G protein. These pathways involve classic second messengers, phosphorylation cascades, lipid signaling, Ca(2+) transients, and gene expression. In addition, IGF-1R triggers signaling in different subcellular locations including the plasma membrane, perinuclear T tubules, and also in internalized vesicles. In this review, we provide a fresh and updated view of the complex IGF-1 scenario in the heart, including a critical focus on therapeutic strategies.


Asunto(s)
Receptor IGF Tipo 1/metabolismo , Calcio/metabolismo , Humanos , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Fosforilación , Transducción de Señal/fisiología , Células Madre/citología
7.
Biochim Biophys Acta ; 1833(12): 3295-3305, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24120520

RESUMEN

Herp is an endoplasmic reticulum (ER) stress inducible protein that participates in the ER-associated protein degradation (ERAD) pathway. However, the contribution of Herp to other protein degradation pathways like autophagy and its connection to other types of stress responses remain unknown. Here we report that Herp regulates autophagy to clear poly-ubiquitin (poly-Ub) protein aggregates. Proteasome inhibition and glucose starvation (GS) led to a high level of poly-Ub protein aggregation that was drastically reduced by stably knocking down Herp (shHerp cells). The enhanced removal of poly-Ub inclusions protected cells from death caused by glucose starvation. Under basal conditions and increasingly after stress, higher LC3-II levels and GFP-LC3 puncta were observed in shHerp cells compared to control cells. Herp knockout cells displayed basal up-regulation of two essential autophagy regulators-Atg5 and Beclin-1, leading to increased autophagic flux. Beclin-1 up-regulation was due to a reduction in Hrd1 dependent proteasomal degradation, and not at transcriptional level. The consequent higher autophagic flux was necessary for the clearance of aggregates and for cell survival. We conclude that Herp operates as a relevant factor in the defense against glucose starvation by modulating autophagy levels. These data may have important implications due to the known up-regulation of Herp in pathological states such as brain and heart ischemia, both conditions associated to acute nutritional stress.


Asunto(s)
Autofagia , Citoprotección , Proteínas de la Membrana/deficiencia , Poliubiquitina/química , Regulación hacia Arriba , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia/efectos de los fármacos , Beclina-1 , Línea Celular , Supervivencia Celular/efectos de los fármacos , Citoprotección/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Glucosa/farmacología , Proteínas de la Membrana/metabolismo , Ratones , Modelos Biológicos , Poliubiquitina/metabolismo , Inhibidores de Proteasoma/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Estructura Cuaternaria de Proteína , Regulación hacia Arriba/efectos de los fármacos
8.
Exp Mol Pathol ; 92(1): 97-104, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22101259

RESUMEN

Increasing evidence indicates that endoplasmic reticulum (ER) stress is involved in various diseases. In the human heart, ischemia/reperfusion has been correlated to ER stress, and several markers of the unfolded protein response (UPR) participate during cardiac remodeling and fibrosis. Here, we used isoproterenol (ISO) injection as a model for in vivo cardiac fibrosis. ISO induced significant cardiomyocyte loss and collagen deposition in the damaged areas of the endocardium. These responses were accompanied by an increase in the protein levels of the luminal ER chaperones BIP and PDI, as well as an increase in the UPR effector CHOP. The use of the chemical chaperone 4-phenylbutyric acid (4-PBA) prevented the activation of the UPR, the increase in luminal chaperones and also, leads to decreased collagen deposition, cardiomyocyte loss into the damaged zones. Our results suggest that cardiac damage and fibrosis induced in vivo by the beta-adrenergic agonist ISO are tightly related to ER stress signaling pathways, and that increasing the ER luminal folding capacity with exogenously administrated 4-PBA is a powerful strategy for preventing the development of cardiac fibrosis. Additionally, 4-PBA might prevent the loss of cardiomyocytes. Our data suggests that the attenuation of ER stress pathways with pharmacological compounds such as the chemical chaperone 4-PBA can prevent the development of cardiac fibrosis and adverse remodeling.


Asunto(s)
Agonistas Adrenérgicos beta/toxicidad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Isoproterenol/toxicidad , Miocardio/patología , Fenilbutiratos/uso terapéutico , Transducción de Señal/efectos de los fármacos , Animales , Fibrosis/inducido químicamente , Fibrosis/patología , Fibrosis/prevención & control , Humanos , Masculino , Chaperonas Moleculares/química , Miocitos Cardíacos/efectos de los fármacos , Pliegue de Proteína , Ratas , Ratas Sprague-Dawley , Factor de Transcripción CHOP/metabolismo
9.
Cardiovasc Res ; 93(2): 320-9, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22135164

RESUMEN

AIMS: Insulin-like growth factor 1 (IGF-1) is known to exert cardioprotective actions. However, it remains unknown if autophagy, a major adaptive response to nutritional stress, contributes to IGF-1-mediated cardioprotection. METHODS AND RESULTS: We subjected cultured neonatal rat cardiomyocytes, as well as live mice, to nutritional stress and assessed cell death and autophagic rates. Nutritional stress induced by serum/glucose deprivation strongly induced autophagy and cell death, and both responses were inhibited by IGF-1. The Akt/mammalian target of rapamycin (mTOR) pathway mediated the effects of IGF-1 upon autophagy. Importantly, starvation also decreased intracellular ATP levels and oxygen consumption leading to AMP-activated protein kinase (AMPK) activation; IGF-1 increased mitochondrial Ca(2+) uptake and mitochondrial respiration in nutrient-starved cells. IGF-1 also rescued ATP levels, reduced AMPK phosphorylation and increased p70(S6K) phosphorylation, which indicates that in addition to Akt/mTOR, IGF-1 inhibits autophagy by the AMPK/mTOR axis. In mice harbouring a liver-specific igf1 deletion, which dramatically reduces IGF-1 plasma levels, AMPK activity and autophagy were increased, and significant heart weight loss was observed in comparison with wild-type starved animals, revealing the importance of IGF-1 in maintaining cardiac adaptability to nutritional insults in vivo. CONCLUSION: Our data support the cardioprotective actions of IGF-1, which, by rescuing the mitochondrial metabolism and the energetic state of cells, reduces cell death and controls the potentially harmful autophagic response to nutritional challenges. IGF-1, therefore, may prove beneficial to mitigate damage induced by excessive nutrient-related stress, including ischaemic disease in multiple tissues.


Asunto(s)
Autofagia/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/farmacología , Miocitos Cardíacos/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/fisiología , Adenosina Trifosfato/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , Ratones , Miocitos Cardíacos/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/fisiología
10.
Mitochondrion ; 12(1): 86-99, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21983689

RESUMEN

Over the past two decades, a complex nuclear transcriptional machinery controlling mitochondrial biogenesis and function has been described. Central to this network are the PGC-1 family coactivators, characterised as master regulators of mitochondrial biogenesis. Recent literature has identified a broader role for PGC-1 coactivators in both cell death and cellular adaptation under conditions of stress, here reviewed in the context of the pathology associated with cancer, neurodegeneration and cardiovascular disease. Moreover, we propose that these studies also imply a novel conceptual framework on the general role of mitochondrial dysfunction in disease. It is now well established that the complex nuclear transcriptional control of mitochondrial biogenesis allows for adaptation of mitochondrial mass and function to environmental conditions. On the other hand, it has also been suggested that mitochondria alter their function according to prevailing cellular energetic requirements and thus function as sensors that generate signals to adjust fundamental cellular processes through a retrograde mitochondria-nucleus signalling pathway. Therefore, altered mitochondrial function can affect cell fate not only directly by modifying cellular energy levels or redox state, but also indirectly, by altering nuclear transcriptional patterns. The current literature on such retrograde signalling in both yeast and mammalian cells is thus reviewed, with an outlook on its potential contribution to disease through the regulation of PGC-1 family coactivators. We propose that further investigation of these pathways will lead to the identification of novel pharmacological targets and treatment strategies to combat disease.


Asunto(s)
Enfermedades Cardiovasculares/fisiopatología , Regulación de la Expresión Génica , Mitocondrias/fisiología , Neoplasias/fisiopatología , Enfermedades Neurodegenerativas/fisiopatología , Estrés Fisiológico , Factores de Transcripción/metabolismo , Animales , Humanos
11.
J Cell Sci ; 124(Pt 13): 2143-52, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21628424

RESUMEN

Increasing evidence indicates that endoplasmic reticulum (ER) stress activates the adaptive unfolded protein response (UPR), but that beyond a certain degree of ER damage, this response triggers apoptotic pathways. The general mechanisms of the UPR and its apoptotic pathways are well characterized. However, the metabolic events that occur during the adaptive phase of ER stress, before the cell death response, remain unknown. Here, we show that, during the onset of ER stress, the reticular and mitochondrial networks are redistributed towards the perinuclear area and their points of connection are increased in a microtubule-dependent fashion. A localized increase in mitochondrial transmembrane potential is observed only in redistributed mitochondria, whereas mitochondria that remain in other subcellular zones display no significant changes. Spatial re-organization of these organelles correlates with an increase in ATP levels, oxygen consumption, reductive power and increased mitochondrial Ca²âº uptake. Accordingly, uncoupling of the organelles or blocking Ca²âº transfer impaired the metabolic response, rendering cells more vulnerable to ER stress. Overall, these data indicate that ER stress induces an early increase in mitochondrial metabolism that depends crucially upon organelle coupling and Ca²âº transfer, which, by enhancing cellular bioenergetics, establishes the metabolic basis for the adaptation to this response.


Asunto(s)
Retículo Endoplásmico/metabolismo , Metabolismo Energético , Mitocondrias/metabolismo , Estrés Fisiológico , Antibacterianos/farmacología , Apoptosis/fisiología , Calcio/metabolismo , Respiración de la Célula , Inhibidores Enzimáticos/farmacología , Células HeLa , Agonistas de los Receptores Histamínicos/farmacología , Humanos , Potencial de la Membrana Mitocondrial , Consumo de Oxígeno/efectos de los fármacos , Fosfatos de Fosfatidilinositol/metabolismo , Transducción de Señal/fisiología
12.
Biochim Biophys Acta ; 1812(1): 23-31, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20637865

RESUMEN

Autophagy is a physiological degradative process key to cell survival during nutrient deprivation, cell differentiation and development. It plays a major role in the turnover of damaged macromolecules and organelles, and it has been involved in the pathogenesis of different cardiovascular diseases. Activation of the adrenergic system is commonly associated with cardiac fibrosis and remodeling, and cardiac fibroblasts are key players in these processes. Whether adrenergic stimulation modulates cardiac fibroblast autophagy remains unexplored. In the present study, we aimed at this question and evaluated the effects of b(2)-adrenergic stimulation upon autophagy. Cultured adult rat cardiac fibroblasts were treated with agonists or antagonists of beta-adrenergic receptors (b-AR), and autophagy was assessed by electron microscopy, GFP-LC3 subcellular distribution, and immunowesternblot of endogenous LC3. The predominant expression of b(2)-ARs was determined and characterized by radioligand binding assays using [(3)H]dihydroalprenolol. Both, isoproterenol and norepinephrine (non-selective b-AR agonists), as well as salbutamol (selective b(2)-AR agonist) increased autophagic flux, and these effects were blocked by propanolol (b-AR antagonist), ICI-118,551 (selective b(2)-AR antagonist), 3-methyladenine but not by atenolol (selective b(1)-AR antagonist). The increase in autophagy was correlated with an enhanced degradation of collagen, and this effect was abrogated by the inhibition of autophagic flux. Overall, our data suggest that b(2)-adrenergic stimulation triggers autophagy in cardiac fibroblasts, and that this response could contribute to reduce the deleterious effects of high adrenergic stimulation upon cardiac fibrosis.


Asunto(s)
Autofagia/fisiología , Colágeno/metabolismo , Fibroblastos/fisiología , Receptores Adrenérgicos beta 2/fisiología , Adenina/análogos & derivados , Adenina/farmacología , Agonistas Adrenérgicos beta/farmacología , Antagonistas Adrenérgicos beta/farmacología , Animales , Autofagia/efectos de los fármacos , Western Blotting , Células Cultivadas , Dihidroalprenolol/metabolismo , Relación Dosis-Respuesta a Droga , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , Isoproterenol/farmacología , Masculino , Microscopía Electrónica de Transmisión , Miocardio/citología , Norepinefrina/farmacología , Propanolaminas/farmacología , Ensayo de Unión Radioligante , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos beta 2/metabolismo , Tritio
13.
Cell Calcium ; 47(2): 112-21, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20097418

RESUMEN

Substantial progress has been made throughout the last decades in the elucidation of the key players and mechanisms responsible for Ca2+ signal generation in both excitable and non-excitable cells. Importantly, these studies led also to the recognition that a close correlation exists between the deregulation of cellular Ca2+ homeostasis and the development of several human pathologies, including neurodegenerative disease. Notwithstanding this advances, much less is certain about the targets and mechanisms by which compromised Ca2+ signaling exerts its effects on cell function and survival. Recently it has been proposed that deregulation of cellular energy metabolism and protein turnover (synthesis, folding and degradation) are also fundamental pathomechanisms of neurodegenerative disease, pointing to the pivotal role of autophagy, a major cellular pathway controlling metabolic homeostasis. Indeed, activation of autophagy has been shown to represent a highly successful strategy to restore normal neuronal function in a variety of models of neurodegenerative disease. Here we review recent advances in elucidating Ca2+ regulation of autophagy and will highlight its relationship to neurodegeneration.


Asunto(s)
Autofagia , Señalización del Calcio , Calcio/metabolismo , Animales , Metabolismo Energético , Humanos , Enfermedades Neurodegenerativas , Complejo de la Endopetidasa Proteasomal/metabolismo
14.
Biochim Biophys Acta ; 1793(9): 1524-32, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19371598

RESUMEN

Autophagy constitutes one of the major responses to stress in eukaryotic cells, and is regulated by a complex network of signaling cascades. Not surprisingly, autophagy is implicated in multiple pathological processes, including infection by pathogens, inflammatory bowel disease, neurodegeneration and cancer. Both oncogenesis and tumor survival are influenced by perturbations of the molecular machinery that controls autophagy. Numerous oncoproteins, including phosphatidylinositol 3-kinase, Akt1 and anti-apoptotic members of the Bcl-2 family suppress autophagy. Conversely, several tumor suppressor proteins (e.g., Atg4c; beclin 1; Bif-1; BH3-only proteins; death-associated protein kinase 1; LKB1/STK11; PTEN; UVRAG) promote the autophagic pathway. This does not entirely apply to p53, one of the most important tumor suppressor proteins, which regulates autophagy in an ambiguous fashion, depending on its subcellular localization. Irrespective of the controversial role of p53, basal levels of autophagy appear to inhibit tumor development. On the contrary, chemotherapy- and metabolic stress-induced activation of the autophagic pathway reportedly contribute to the survival of formed tumors, thereby favoring resistance. In this context, autophagy inhibition would represent a major therapeutic target for chemosensitization. Here, we will review the current knowledge on the dual role of autophagy as an anti- and pro-tumor mechanism.


Asunto(s)
Autofagia , Citoprotección , Neoplasias/patología , Animales , Humanos , Neoplasias/terapia , Proteína p53 Supresora de Tumor/metabolismo
15.
Apoptosis ; 14(2): 182-90, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19152031

RESUMEN

QM31 represents a new class of cytoprotective agents that inhibit the formation of the apoptosome, the caspase activation complex composed by Apaf-1, cytochrome c, dATP and caspase-9. Here, we analyzed the cellular effects of QM31, as compared to the prototypic caspase inhibitor Z-VAD-fmk. QM31 was as efficient as Z-VAD-fmk in suppressing caspase-3 activation, and conferred a similar cytoprotective effect. In contrast to Z-VAD-fmk, QM31 inhibited the release of cytochrome c from mitochondria, an unforeseen property that may contribute to its pronounced cytoprotective activity. Moreover, QM31 suppressed the Apaf-1-dependent intra-S-phase DNA damage checkpoint. These results suggest that QM31 can interfere with the two known functions of Apaf-1, namely apoptosome assembly/activation and intra-S-phase cell cycle arrest. Moreover, QM31 can inhibit mitochondrial outer membrane permeabilization, an effect that is independent from its action on Apaf-1.


Asunto(s)
Factor Apoptótico 1 Activador de Proteasas/antagonistas & inhibidores , Azepinas/farmacología , Daño del ADN , Mitocondrias/efectos de los fármacos , Peptoides/farmacología , Fase S/efectos de los fármacos , Clorometilcetonas de Aminoácidos/farmacología , Factor Apoptótico 1 Activador de Proteasas/deficiencia , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Permeabilidad de la Membrana Celular/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Humanos , Neoplasias Pulmonares/patología , ARN Interferente Pequeño/metabolismo , Transfección
16.
Cell Cycle ; 7(19): 3056-61, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18818522

RESUMEN

The knockout, knockdown or chemical inhibition of p53 stimulates autophagy. Moreover, autophagy-inducing stimuli such as nutrient depletion, rapamycin or lithium cause the depletion of cytoplasmic p53, which in turn is required for the induction of autophagy. Here, we show that retransfection of p53(-/-) HCT 116 colon carcinoma cells with wild type p53 decreases autophagy down to baseline levels. Surprisingly, one third among a panel of 22 cancer-associated p53 single amino acid mutants also inhibited autophagy when transfected into p53(-/-) cells. Those variants of p53 that preferentially localize to the cytoplasm effectively repressed autophagy, whereas p53 mutants that display a prominently nuclear distribution failed to inhibit autophagy. The investigation of a series of deletion mutants revealed that removal of the DNA-binding domain from p53 fails to interfere with its role in the regulation of autophagy. Altogether, these results identify the cytoplasmic localization of p53 as the most important feature for p53-mediated autophagy inhibition. Moreover, the structural requirements for the two biological activities of extranuclear p53, namely induction of apoptosis and inhibition of autophagy, are manifestly different.


Asunto(s)
Autofagia/genética , Citoplasma/metabolismo , Mutación , Proteína p53 Supresora de Tumor/análisis , Proteína p53 Supresora de Tumor/genética , Células HCT116 , Humanos , Proteína p53 Supresora de Tumor/metabolismo
17.
Biochem Soc Trans ; 36(Pt 5): 786-90, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18793137

RESUMEN

Macroautophagy, often referred to as autophagy, designates the process by which portions of the cytoplasm, intracellular organelles and long-lived proteins are engulfed in double-membraned vacuoles (autophagosomes) and sent for lysosomal degradation. Basal levels of autophagy contribute to the maintenance of intracellular homoeostasis by ensuring the turnover of supernumerary, aged and/or damaged components. Under conditions of starvation, the autophagic pathway operates to supply cells with metabolic substrates, and hence represents an important pro-survival mechanism. Moreover, autophagy is required for normal development and for the protective response to intracellular pathogens. Conversely, uncontrolled autophagy is associated with a particular type of cell death (termed autophagic, or type II) that is characterized by the massive accumulation of autophagosomes. Regulators of apoptosis (e.g. Bcl-2 family members) also modulate autophagy, suggesting an intimate cross-talk between these two degradative pathways. It is still unclear whether autophagic vacuolization has a causative role in cell death or whether it represents the ultimate attempt of cells to cope with lethal stress. For a multicellular organism, autophagic cell death might well represent a pro-survival mechanism, by providing metabolic supplies during whole-body nutrient deprivation. Alternatively, type II cell death might contribute to the disposal of cell corpses when heterophagy is deficient. Here, we briefly review the roles of autophagy in cell death and its avoidance.


Asunto(s)
Autofagia/fisiología , Muerte Celular/fisiología , Animales , Supervivencia Celular , Humanos
18.
Nat Cell Biol ; 10(6): 676-87, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18454141

RESUMEN

Multiple cellular stressors, including activation of the tumour suppressor p53, can stimulate autophagy. Here we show that deletion, depletion or inhibition of p53 can induce autophagy in human, mouse and nematode cells subjected to knockout, knockdown or pharmacological inhibition of p53. Enhanced autophagy improved the survival of p53-deficient cancer cells under conditions of hypoxia and nutrient depletion, allowing them to maintain high ATP levels. Inhibition of p53 led to autophagy in enucleated cells, and cytoplasmic, not nuclear, p53 was able to repress the enhanced autophagy of p53(-/-) cells. Many different inducers of autophagy (for example, starvation, rapamycin and toxins affecting the endoplasmic reticulum) stimulated proteasome-mediated degradation of p53 through a pathway relying on the E3 ubiquitin ligase HDM2. Inhibition of p53 degradation prevented the activation of autophagy in several cell lines, in response to several distinct stimuli. These results provide evidence of a key signalling pathway that links autophagy to the cancer-associated dysregulation of p53.


Asunto(s)
Autofagia , Citoplasma/metabolismo , Regulación de la Expresión Génica , Genes p53 , Proteína p53 Supresora de Tumor/fisiología , Animales , Línea Celular Tumoral , Retículo Endoplásmico/metabolismo , Humanos , Hipoxia , Lisosomas/metabolismo , Ratones , Ratones Transgénicos , Modelos Biológicos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
19.
Gerontology ; 54(2): 92-9, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18451641

RESUMEN

Many features of aging result from the incapacity of cells to adapt to stress conditions. When damage accumulates irreversibly, mitotic cells from renewable tissues rely on either of two mechanisms to avoid replication. They can permanently arrest the cell cycle (cellular senescence) or trigger cell death programs. Apoptosis (self-killing) is the best-described form of programmed cell death, but autophagy (self-eating), which is a lysosomal degradation pathway essential for homeostasis, reportedly contributes to cell death as well. Unlike mitotic cells, postmitotic cells like neurons or cardiomyocytes cannot become senescent since they are already terminally differentiated. The fate of these cells entirely depends on their ability to cope with stress. Autophagy then operates as a major homeostatic mechanism to eliminate damaged organelles, long-lived or aberrant proteins and superfluous portions of the cytoplasm. In this mini-review, we briefly summarize the molecular networks that allow damaged cells either to adapt to stress or to engage in programmed-cell-death pathways.


Asunto(s)
Apoptosis/fisiología , Autofagia/fisiología , Senescencia Celular/fisiología , Envejecimiento/fisiología , Animales
20.
Curr Mol Med ; 8(2): 78-91, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18336289

RESUMEN

Macroautophagy (commonly referred to as autophagy) is the process by which intact organelles and/or large portions of the cytoplasm are engulfed within double-membraned autophagic vacuoles for degradation. Whereas basal levels of autophagy ensure the physiological turnover of old and damaged organelles, the massive accumulation of autophagic vacuoles may represent either an alternative pathway of cell death or an ultimate attempt for cells to survive by adapting to stress. The activation of the autophagic pathway beyond a certain threshold may promote cell death directly, by causing the collapse of cellular functions as a result of cellular atrophy (autophagic, or type II, cell death). Alternatively, autophagy can lead to the execution of apoptotic (type I) or necrotic (type III) cell death programs, presumably via common regulators such as proteins from the Bcl-2 family. On the other hand, limited self-eating can provide cells with metabolic substrates to meet their energetic demands under stressful conditions, such as nutrient deprivation, or favor the selective elimination of damaged (and potentially dangerous) organelles. In these instances, autophagy operates as a pro-survival mechanism. The coordinate regulation of these opposite effects of autophagy relies upon a complex network of signal transducers, most of which also participate in non-autophagic signaling cascades. Thus, autophagy occupies a crucial position within the cell's metabolism, and its modulation may represent an alternative therapeutic strategy in several pathological settings including cancer and neurodegeneration. Here, we present a general outline of autophagy followed by a detailed analysis of organelle-specific autophagic pathways and of their intimate connections with cell death.


Asunto(s)
Apoptosis/fisiología , Autofagia/fisiología , Animales , Humanos , Modelos Biológicos , Orgánulos/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...