Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Am J Physiol Endocrinol Metab ; 303(2): E172-9, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22535743

RESUMEN

Poly(ADP)-ribose polymerase (PARP) is an abundant nuclear protein that is activated by DNA damage; once active, it modifies nuclear proteins through attachment of poly(ADP)-ribose units derived from ß-nicotinamide adenine dinucleotide (NAD(+)). In mice, the deletion of PARP-1 attenuates tissue injury in a number of animal models of human disease, including streptozotocin-induced diabetes. Also, inflammatory cell signaling and inflammatory gene expression are attenuated in macrophages isolated from endotoxin-treated PARP-1-deficient mice. In this study, the effects of PARP-1 deletion on cytokine-mediated ß-cell damage and macrophage activation were evaluated. There are no defects in inflammatory mediator signaling or inflammatory gene expression in macrophages and islets isolated from PARP-1-deficient mice. While PARP-1 deficiency protects islets against cytokine-induced islet cell death as measured by biochemical assays of membrane polarization, the genetic absence of PARP-1 does not effect cytokine-induced inhibition of insulin secretion or cytokine-induced DNA damage in islets. While PARP-1 deficiency appears to provide protection from cell death, it fails to provide protection against the inhibitory actions of cytokines on insulin secretion or the damaging actions on islet DNA integrity.


Asunto(s)
Citocinas/metabolismo , Células Secretoras de Insulina/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Animales , Apoptosis , Células Cultivadas , Daño del ADN , Femenino , Eliminación de Gen , Expresión Génica , Insulina/metabolismo , Secreción de Insulina , Activación de Macrófagos , Macrófagos/metabolismo , Masculino , Potenciales de la Membrana , Ratones , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/genética , Transducción de Señal
2.
3.
Mol Cell Biol ; 31(21): 4286-97, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21896783

RESUMEN

While there can be detrimental consequences of nitric oxide production at pathological concentrations, eukaryotic cells have evolved protective mechanisms to defend themselves against this damage. The unfolded-protein response (UPR), activated by misfolded proteins and oxidative stress, is one adaptive mechanism that is employed to protect cells from stress. Nitric oxide is a potent activator of AMP-activated protein kinase (AMPK), and AMPK participates in the cellular defense against nitric oxide-mediated damage in pancreatic ß-cells. In this study, the mechanism of AMPK activation by nitric oxide was explored. The known AMPK kinases LKB1, CaMKK, and TAK1 are not required for the activation of AMPK by nitric oxide. Instead, this activation is dependent on the endoplasmic reticulum (ER) stress-activated protein IRE1. Nitric oxide-induced AMPK phosphorylation and subsequent signaling to AMPK substrates, including Raptor, acetyl coenzyme A carboxylase, and PGC-1α, is attenuated in IRE1α-deficient cells. The endoribonuclease activity of IRE1 appears to be required for AMPK activation in response to nitric oxide. In addition to nitric oxide, stimulation of IRE1 endoribonuclease activity with the flavonol quercetin leads to IRE1-dependent AMPK activation. These findings indicate that the RNase activity of IRE1 participates in AMPK activation and subsequent signaling through multiple AMPK-dependent pathways in response to nitrosative stress.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Endorribonucleasas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/genética , Animales , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Línea Celular , Estrés del Retículo Endoplásmico , Endorribonucleasas/deficiencia , Endorribonucleasas/genética , Activación Enzimática/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Modelos Biológicos , Complejos Multiproteicos , Óxido Nítrico/farmacología , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteínas/antagonistas & inhibidores , ARN Interferente Pequeño/genética , Ratas , Transducción de Señal , Serina-Treonina Quinasas TOR
4.
J Biol Chem ; 286(10): 8338-8348, 2011 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-21196578

RESUMEN

For many cell types, including pancreatic ß-cells, nitric oxide is a mediator of cell death; paradoxically, nitric oxide can also activate pathways that promote the repair of cellular damage. In this report, a role for FoxO1-dependent transcriptional activation and its regulation by SIRT1 in determining the cellular response to nitric oxide is provided. In response to nitric oxide, FoxO1 translocates from the cytoplasm to the nucleus and stimulates the expression of the DNA repair gene GADD45α, resulting in FoxO1-dependent DNA repair. FoxO1-dependent gene expression appears to be regulated by the NAD(+)-dependent deacetylase SIRT1. In response to SIRT1 inhibitors, the FoxO1-dependent protective actions of nitric oxide (GADD45α expression and DNA repair) are attenuated, and FoxO1 activates a proapoptotic program that includes PUMA (p53-up-regulated mediator of apoptosis) mRNA accumulation and caspase-3 cleavage. These findings support primary roles for FoxO1 and SIRT1 in regulating the cellular responses of ß-cells to nitric oxide.


Asunto(s)
Núcleo Celular/metabolismo , Reparación del ADN , Factores de Transcripción Forkhead/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Óxido Nítrico/metabolismo , Sirtuina 1/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Caspasa 3/genética , Caspasa 3/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Núcleo Celular/genética , Factores Relajantes Endotelio-Dependientes/metabolismo , Factores Relajantes Endotelio-Dependientes/farmacología , Factores de Transcripción Forkhead/genética , Masculino , Proteínas del Tejido Nervioso/genética , Óxido Nítrico/farmacología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Sirtuina 1/genética
5.
Tissue Eng Part A ; 17(3-4): 399-406, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20807014

RESUMEN

Peptide amphiphile (PA) is a peptide-based biomaterial that can self-assemble into a nanostructured gel-like scaffold, mimicking the chemical and biological complexity of natural extracellular matrix. To evaluate the capacity of the PA scaffold to improve islet function and survival in vitro, rat islets were cultured in three different groups--(1) bare group: isolated rat islets cultured in a 12-well nontissue culture-treated plate; (2) insert group: isolated rat islets cultured in modified insert chambers; (3) nanomatrix group: isolated rat islets encapsulated within the PA nanomatrix gel and cultured in modified insert chambers. Over 14 days, both the bare and insert groups showed a marked decrease in insulin secretion, whereas the nanomatrix group maintained glucose-stimulated insulin secretion. Moreover, entire islets in the nanomatrix gel stained positive for dithizone up to 14 days, indicating better maintained glucose-stimulated insulin production. Fluorescein diacetate/propidium iodide staining results also verified necrosis in the bare and insert groups after 7 days, whereas the PA nanomatrix gel maintained islet viability after 14 days. Thus, these results demonstrate the potential of PAs as an intermediary scaffold for increasing the efficacy of pancreatic islet transplantation.


Asunto(s)
Materiales Biomiméticos/síntesis química , Matriz Extracelular/química , Trasplante de Islotes Pancreáticos/fisiología , Nanoestructuras/química , Páncreas Artificial , Animales , Proliferación Celular , Supervivencia Celular , Geles/química , Masculino , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley
6.
J Biol Chem ; 285(5): 3191-200, 2010 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-19933272

RESUMEN

During the initial autoimmune response in type 1 diabetes, islets are exposed to a damaging mix of pro-inflammatory molecules that stimulate the production of nitric oxide by beta-cells. Nitric oxide causes extensive but reversible cellular damage. In response to nitric oxide, the cell activates pathways for functional recovery and adaptation as well as pathways that direct beta-cell death. The molecular events that dictate cellular fate following nitric oxide-induced damage are currently unknown. In this study, we provide evidence that AMPK plays a primary role controlling the response of beta-cells to nitric oxide-induced damage. AMPK is transiently activated by nitric oxide in insulinoma cells and rat islets following IL-1 treatment or by the exogenous addition of nitric oxide. Active AMPK promotes the functional recovery of beta-cell oxidative metabolism and abrogates the induction of pathways that mediate cell death such as caspase-3 activation following exposure to nitric oxide. Overall, these data show that nitric oxide activates AMPK and that active AMPK suppresses apoptotic signaling allowing the beta-cell to recover from nitric oxide-mediated cellular stress.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Células Secretoras de Insulina/patología , Óxido Nítrico/metabolismo , Aconitato Hidratasa/metabolismo , Animales , Caspasa 3/metabolismo , Muerte Celular , Linaje de la Célula , Ensayo Cometa , Células Secretoras de Insulina/metabolismo , Insulinoma , Interleucina-1/metabolismo , Masculino , Nitritos/química , Ratas , Ratas Sprague-Dawley
7.
Am J Physiol Endocrinol Metab ; 297(5): E1187-96, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19738038

RESUMEN

For many cell types, including pancreatic ß-cells, nitric oxide is a mediator of cell death; however, it is paradoxical that for a given cell type nitric oxide can induce both necrosis and apoptosis. This report tests the hypothesis that cell death mediated by nitric oxide shifts from an early necrotic to a late apoptotic event. Central to this transition is the ability of ß-cells to respond and repair nitric oxide-mediated damage. ß-Cells have the ability to repair DNA that is damaged following 24-h incubation with IL-1; however, cytokine-induced DNA damage becomes irreversible following 36-h incubation. This irreversible DNA damage following 36-h incubation with IL-1 correlates with the activation of caspase-3 (cleavage and activity). The increase in caspase activity correlates with reductions in endogenous nitric oxide production, as nitric oxide is an inhibitor of caspase activity. In contrast, caspase cleavage or activation is not observed under conditions in which ß-cells are capable of repairing damaged DNA (24-h incubation with cytokines). These findings provide evidence that ß-cell death in response to cytokines shifts from an early necrotic process to apoptosis and that this shift is associated with irreversible DNA damage and caspase-3 activation.


Asunto(s)
Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Citocinas/farmacología , Daño del ADN/fisiología , Células Secretoras de Insulina/efectos de los fármacos , Óxido Nítrico/farmacología , Animales , Caspasa 3/metabolismo , Separación Celular , Ensayo Cometa , Reparación del ADN/efectos de los fármacos , Metabolismo Energético/fisiología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Humanos , Inmunohistoquímica , Técnicas In Vitro , Interleucina-1/antagonistas & inhibidores , Interleucina-1/farmacología , Masculino , Necrosis , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa
8.
J Biol Chem ; 284(40): 27402-8, 2009 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-19648647

RESUMEN

Proinflammatory cytokines induce nitric oxide-dependent DNA damage and ultimately beta-cell death. Not only does nitric oxide cause beta-cell damage, it also activates a functional repair process. In this study, the mechanisms activated by nitric oxide that facilitate the repair of damaged beta-cell DNA are examined. JNK plays a central regulatory role because inhibition of this kinase attenuates the repair of nitric oxide-induced DNA damage. p53 is a logical target of JNK-dependent DNA repair; however, nitric oxide does not stimulate p53 activation or accumulation in beta-cells. Further, knockdown of basal p53 levels does not affect DNA repair. In contrast, expression of growth arrest and DNA damage (GADD) 45alpha, a DNA repair gene that can be regulated by p53-dependent and p53-independent pathways, is stimulated by nitric oxide in a JNK-dependent manner, and knockdown of GADD45alpha expression attenuates the repair of nitric oxide-induced beta-cell DNA damage. These findings show that beta-cells have the ability to repair nitric oxide-damaged DNA and that JNK and GADD45alpha mediate the p53-independent repair of this DNA damage.


Asunto(s)
Proteínas de Ciclo Celular/genética , Daño del ADN/efectos de los fármacos , Reparación del ADN , Regulación de la Expresión Génica , Células Secretoras de Insulina/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Óxido Nítrico/farmacología , Proteínas Nucleares/genética , Animales , Células Cultivadas , Activación Enzimática , Femenino , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/enzimología , Ratas , Ratas Sprague-Dawley , Proteína p53 Supresora de Tumor/metabolismo
9.
Apoptosis ; 11(11): 1933-44, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17013754

RESUMEN

The effect of Zn on p53-independent cell death was examined in IIC9 embryonic fibroblasts. Despite the fact that these cells are p53-minus, Zn-mediated death occurs via an apoptotic mechanism. Death is facilitated by the presence of the Zn ionophore, pyrithione, indicating that intracellular Zn initiates the death response. Our investigations of the mechanism of Zn action demonstrate that Zn induces the death of IIC9 cells in a manner that is ERK-dependent. Expression of dn-(dominant negative)Ras attenuates ERK1/2 activation by Zn, and correspondingly reduces its cytotoxic effects. Raf-RBD pull-down experiments confirm that Zn treatment activates Ras and identified H-Ras as the specific isoform activated. This contrasts the activation of N-Ras that occurs when IIC9 cells are stimulated with thrombin. Thus, although the prolonged activation of the Ras/ERK pathway by Zn is similar to that seen when induced by mitogen, the distinguishing feature appears to be the isoform specificity of Ras activation.


Asunto(s)
Apoptosis , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Zinc/metabolismo , Animales , Apoptosis/efectos de los fármacos , Cloruros/toxicidad , Cricetinae , Embrión de Mamíferos/citología , Fibroblastos/citología , Genes ras , Ionóforos/farmacología , Isoformas de Proteínas/metabolismo , Piridinas/farmacología , Tionas/farmacología , Transfección , Compuestos de Zinc/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA