Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Aging ; 4(1): 48-61, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38057386

RESUMEN

Transient events during development can exert long-lasting effects on organismal lifespan. Here we demonstrate that exposure of Caenorhabditis elegans to reactive oxygen species during development protects against amyloid-induced proteotoxicity later in life. We show that this protection is initiated by the inactivation of the redox-sensitive H3K4me3-depositing COMPASS complex and conferred by a substantial increase in the heat-shock-independent activity of heat shock factor 1 (HSF-1), a longevity factor known to act predominantly during C. elegans development. We show that depletion of HSF-1 leads to marked rearrangements of the organismal lipid landscape and a significant decrease in mitochondrial ß-oxidation and that both lipid and metabolic changes contribute to the protective effects of HSF-1 against amyloid toxicity. Together, these findings link developmental changes in the histone landscape, HSF-1 activity and lipid metabolism to protection against age-associated amyloid toxicities later in life.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Factores de Transcripción/genética , Histonas/metabolismo , Metabolismo de los Lípidos/genética , Proteínas Amiloidogénicas/metabolismo , Calidad de Vida , Lípidos/farmacología
2.
J Biol Chem ; 299(3): 102994, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36773802

RESUMEN

Nitric oxide (NO) plays a dual role in regulating DNA damage response (DDR) signaling in pancreatic ß-cells. As a genotoxic agent, NO activates two types of DDR signaling; however, when produced at micromolar levels by the inducible isoform of NO synthase, NO inhibits DDR signaling and DDR-induced apoptosis in a ß-cell-selective manner. DDR signaling inhibition by NO correlates with mitochondrial oxidative metabolism inhibition and decreases in ATP and NAD+. Unlike most cell types, ß-cells do not compensate for impaired mitochondrial oxidation by increasing glycolytic flux, and this metabolic inflexibility leads to a decrease in ATP and NAD+. Here, we used multiple analytical approaches to determine changes in intermediary metabolites in ß-cells and non-ß-cells treated with NO or complex I inhibitor rotenone. In addition to ATP and NAD+, glycolytic and tricarboxylic acid cycle intermediates as well as NADPH are significantly decreased in ß-cells treated with NO or rotenone. Consistent with glucose-6-phosphate residing at the metabolic branchpoint for glycolysis and the pentose phosphate pathway (NADPH), we show that mitochondrial oxidation inhibitors limit glucose uptake in a ß-cell-selective manner. Our findings indicate that the ß-cell-selective inhibition of DDR signaling by NO is associated with a decrease in ATP to levels that fall significantly below the KM for ATP of glucokinase (glucose uptake) and suggest that this action places the ß-cell in a state of suspended animation where it is metabolically inert until NO is removed, and metabolic function can be restored.


Asunto(s)
NAD , Óxido Nítrico , Óxido Nítrico/metabolismo , NADP/metabolismo , NAD/metabolismo , Rotenona/farmacología , Daño del ADN , Adenosina Trifosfato/metabolismo , Glucosa/metabolismo
3.
Cell Cycle ; 20(22): 2337-2347, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34657571

RESUMEN

Studies in Caenorhabditis elegans have revealed that even a genetically identical population of animals exposed to the same environment displays a remarkable level of variability in individual lifespan. Stochasticity factors, occurring seemingly by chance or at random, are thought to account for a large part of this variability. Recent studies in our lab using C. elegans now revealed that naturally occurring variations in the levels of reactive oxygen species experienced early in life contribute to the observed lifespan variability, and likely serve as stochasticity factors in aging. Here, we will highlight how developmental events can positively shape lifespan and stress responses via a redox-sensitive epigenetic regulator, and discuss the outstanding questions and future directions on the complex relationship between reactive oxygen species and aging.


Asunto(s)
Proteínas de Caenorhabditis elegans , Longevidad , Envejecimiento/genética , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Histonas , Longevidad/genética , Estrés Oxidativo , Especies Reactivas de Oxígeno
4.
J Biol Chem ; 296: 100388, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33567339

RESUMEN

We have shown that nitric oxide limits ataxia-telangiectasia mutated signaling by inhibiting mitochondrial oxidative metabolism in a ß-cell selective manner. In this study, we examined the actions of nitric oxide on a second DNA damage response transducer kinase, ataxia-telangiectasia and Rad3-related protein (ATR). In ß-cells and non-ß-cells, nitric oxide activates ATR signaling by inhibiting ribonucleotide reductase; however, when produced at inducible nitric oxide synthase-derived (low micromolar) levels, nitric oxide impairs ATR signaling in a ß-cell selective manner. The inhibitory actions of nitric oxide are associated with impaired mitochondrial oxidative metabolism and lack of glycolytic compensation that result in a decrease in ß-cell ATP. Like nitric oxide, inhibitors of mitochondrial respiration reduce ATP levels and limit ATR signaling in a ß-cell selective manner. When non-ß-cells are forced to utilize mitochondrial oxidative metabolism for ATP generation, their response is more like ß-cells, as nitric oxide and inhibitors of mitochondrial respiration attenuate ATR signaling. These studies support a dual role for nitric oxide in regulating ATR signaling. Nitric oxide activates ATR in all cell types examined by inhibiting ribonucleotide reductase, and in a ß-cell selective manner, inducible nitric oxide synthase-derived levels of nitric oxide limit ATR signaling by attenuating mitochondrial oxidative metabolism and depleting ATP.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Daño del ADN , Células Secretoras de Insulina/metabolismo , Mitocondrias/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico/farmacología , Animales , Células Cultivadas , Células Secretoras de Insulina/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Ratas , Transducción de Señal
5.
Biochem Pharmacol ; 176: 113907, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32171728

RESUMEN

Insulin, produced by pancreatic ß-cells, is responsible for the control of whole-body glucose metabolism. Insulin is secreted by pancreatic ß-cells in a tightly regulated process that is controlled by the serum level of glucose, glucose sensing and glucose oxidative metabolism. The regulation of intermediary metabolism in ß-cells is unique as these cells oxidize glucose to CO2 on substrate supply while mitochondrial oxidative metabolism occurs on demand, for the production of intermediates or energy production, in most cell types. This review discusses recent findings that the regulation of intermediary metabolism by nitric oxide attenuates the DNA damage response (DDR) and DNA damage-dependent apoptosis in a ß-cell selective manner. Specific focus is placed on the mechanisms by which iNOS derived nitric oxide (low micromolar levels) regulates DDR activation via the inhibition of intermediary metabolism. The physiological significance of the association of metabolism, nitric oxide and DDR signaling for cancer biology and diabetes is discussed.


Asunto(s)
Daño del ADN , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animales , Diabetes Mellitus/metabolismo , Humanos , Neoplasias/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo
6.
J Biol Chem ; 295(8): 2385-2397, 2020 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-31915247

RESUMEN

Viral infection is an environmental trigger that has been suggested to initiate pancreatic ß-cell damage, leading to the development of autoimmune diabetes. Viruses potently activate the immune system and can damage ß cells by either directly infecting them or stimulating the production of secondary effector molecules (such as proinflammatory cytokines) during bystander activation. However, how and where ß cells recognize viruses is unclear, and the antiviral responses that are initiated following virus recognition are incompletely understood. In this study, we show that the ß-cell response to dsRNA, a viral replication intermediate known to activate antiviral responses, is determined by the cellular location of sensing (intracellular versus extracellular) and differs from the cellular response to cytokine treatment. Using biochemical and immunological methods, we show that ß cells selectively respond to intracellular dsRNA by expressing type I interferons (IFNs) and inducing apoptosis, but that they do not respond to extracellular dsRNA. These responses differ from the activities of cytokines on ß cells, which are mediated by inducible nitric oxide synthase expression and ß-cell production of nitric oxide. These findings provide evidence that the antiviral activities of type I IFN production and apoptosis are elicited in ß cells via the recognition of intracellular viral replication intermediates and that ß cells lack the capacity to respond to extracellular viral intermediates known to activate innate immune responses.


Asunto(s)
Células Secretoras de Insulina/virología , ARN Bicatenario/metabolismo , Animales , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Supervivencia Celular/efectos de los fármacos , Daño del ADN , Activación Enzimática/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/patología , Interferón Tipo I/metabolismo , Masculino , Óxido Nítrico Sintasa de Tipo II/metabolismo , Poli I-C/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 3/metabolismo
7.
Nature ; 576(7786): 301-305, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31801997

RESUMEN

A central aspect of aging research concerns the question of when individuality in lifespan arises1. Here we show that a transient increase in reactive oxygen species (ROS), which occurs naturally during early development in a subpopulation of synchronized Caenorhabditis elegans, sets processes in motion that increase stress resistance, improve redox homeostasis and ultimately prolong lifespan in those animals. We find that these effects are linked to the global ROS-mediated decrease in developmental histone H3K4me3 levels. Studies in HeLa cells confirmed that global H3K4me3 levels are ROS-sensitive and that depletion of H3K4me3 levels increases stress resistance in mammalian cell cultures. In vitro studies identified SET1/MLL histone methyltransferases as redox sensitive units of the H3K4-trimethylating complex of proteins (COMPASS). Our findings implicate a link between early-life events, ROS-sensitive epigenetic marks, stress resistance and lifespan.


Asunto(s)
Longevidad , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Animales , Caenorhabditis elegans , Regulación hacia Abajo , Histonas/metabolismo , Larva
8.
Mol Cell Biol ; 39(18)2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31235477

RESUMEN

In this report, we show that nitric oxide suppresses DNA damage response (DDR) signaling in the pancreatic ß-cell line INS 832/13 and rat islets by inhibiting intermediary metabolism. Nitric oxide is known to inhibit complex IV of the electron transport chain and aconitase of the Krebs cycle. Non-ß cells compensate by increasing glycolytic metabolism to maintain ATP levels; however, ß cells lack this metabolic flexibility, resulting in a nitric oxide-dependent decrease in ATP and NAD+ Like nitric oxide, mitochondrial toxins inhibit DDR signaling in ß cells by a mechanism that is associated with a decrease in ATP. Non-ß cells compensate for the effects of mitochondrial toxins with an adaptive shift to glycolytic ATP generation that allows for DDR signaling. Forcing non-ß cells to derive ATP via mitochondrial respiration (replacing glucose with galactose in the medium) and glucose deprivation sensitizes these cells to nitric oxide-mediated inhibition of DDR signaling. These findings indicate that metabolic flexibility is necessary to maintain DDR signaling under conditions in which mitochondrial oxidative metabolism is inhibited and support the inhibition of oxidative metabolism (decreased ATP) as one protective mechanism by which nitric oxide attenuates DDR-dependent ß-cell apoptosis.


Asunto(s)
Reparación del ADN/efectos de los fármacos , Glucólisis/efectos de los fármacos , Células Secretoras de Insulina/citología , Óxido Nítrico/farmacología , Adenosina Trifosfato/metabolismo , Animales , Línea Celular , Respiración de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Daño del ADN , Células Hep G2 , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , NAD/metabolismo , Ratas , Ratas Sprague-Dawley
9.
J Biol Chem ; 294(13): 4843-4853, 2019 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-30659092

RESUMEN

Oxidative stress is thought to promote pancreatic ß-cell dysfunction and contribute to both type 1 and type 2 diabetes. Reactive oxygen species (ROS), such as superoxide and hydrogen peroxide, are mediators of oxidative stress that arise largely from electron leakage during oxidative phosphorylation. Reports that ß-cells express low levels of antioxidant enzymes, including catalase and GSH peroxidases, have supported a model in which ß-cells are ill-equipped to detoxify ROS. This hypothesis seems at odds with the essential role of ß-cells in the control of metabolic homeostasis and organismal survival through exquisite coupling of oxidative phosphorylation, a prominent ROS-producing pathway, to insulin secretion. Using glucose oxidase to deliver H2O2 continuously over time and Amplex Red to measure extracellular H2O2 concentration, we found here that ß-cells can remove micromolar levels of this oxidant. This detoxification pathway utilizes the peroxiredoxin/thioredoxin antioxidant system, as selective chemical inhibition or siRNA-mediated depletion of thioredoxin reductase sensitized ß-cells to continuously generated H2O2 In contrast, when delivered as a bolus, H2O2 induced the DNA damage response, depleted cellular energy stores, and decreased ß-cell viability independently of thioredoxin reductase inhibition. These findings show that ß-cells have the capacity to detoxify micromolar levels of H2O2 through a thioredoxin reductase-dependent mechanism and are not as sensitive to oxidative damage as previously thought.


Asunto(s)
Peróxido de Hidrógeno/metabolismo , Células Secretoras de Insulina/metabolismo , Peroxirredoxinas/metabolismo , Tiorredoxinas/metabolismo , Animales , Supervivencia Celular , Daño del ADN , Secreción de Insulina , Masculino , Oxidación-Reducción , Ratas , Ratas Sprague-Dawley , Reductasa de Tiorredoxina-Disulfuro/metabolismo
10.
Diabetes ; 67(5): 898-910, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29444892

RESUMEN

Nitric oxide is produced at micromolar levels by pancreatic ß-cells during exposure to proinflammatory cytokines. While classically viewed as damaging, nitric oxide also activates pathways that promote ß-cell survival. We have shown that nitric oxide, in a cell type-selective manner, inhibits the DNA damage response (DDR) and, in doing so, protects ß-cells from DNA damage-induced apoptosis. This study explores potential mechanisms by which nitric oxide inhibits DDR signaling. We show that inhibition of DDR signaling (measured by γH2AX formation and the phosphorylation of KAP1) is selective for nitric oxide, as other forms of reactive oxygen/nitrogen species do not impair DDR signaling. The kinetics and broad range of DDR substrates that are inhibited suggest that protein phosphatase activation may be one mechanism by which nitric oxide attenuates DDR signaling in ß-cells. While protein phosphatase 1 (PP1) is a primary regulator of DDR signaling and an inhibitor of PP1 (IPP1) is selectively expressed only in ß-cells, disruption of either IPP1 or PP1 does not modify the inhibitory actions of nitric oxide on DDR signaling in ß-cells. These findings support a PP1-independent mechanism by which nitric oxide selectively impairs DDR signaling and protects ß-cells from DNA damage-induced apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Daño del ADN , Células Secretoras de Insulina/efectos de los fármacos , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico/farmacología , Proteína Fosfatasa 1/efectos de los fármacos , Proteínas/efectos de los fármacos , Animales , Supervivencia Celular , Histonas/efectos de los fármacos , Histonas/metabolismo , Células Secretoras de Insulina/metabolismo , Fosfoproteínas/efectos de los fármacos , Fosfoproteínas/metabolismo , Fosforilación , Proteína Fosfatasa 1/metabolismo , Proteínas/metabolismo , Ratas , Transducción de Señal
11.
Mol Cell Biol ; 38(8)2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29378831

RESUMEN

Palmitate attenuates insulin secretion and reduces the viability of insulin-producing cells. Previous studies identified the aberrant palmitoylation or mispalmitoylation of proteins as one mechanism by which palmitate causes ß-cell damage. In this report, we identify a role for lysosomal protein degradation as a mechanism by which ß cells defend themselves against excess palmitate. The cation-independent mannose 6-phosphate receptor (CI-MPR) is responsible for the trafficking of mannose 6-phosphate-tagged proteins to lysosomes via Golgi sorting and from extracellular locations through endocytosis. RINm5F cells, which are highly sensitive to palmitate, lack CI-MPR. The reconstitution of CI-MPR expression attenuates the induction of endoplasmic reticulum (ER) stress and the toxic effects of palmitate on RINm5F cell viability. INS832/13 cells express CI-MPR and are resistant to the palmitate-mediated loss of cell viability. The reduction of CI-MPR expression increases the sensitivity of INS832/13 cells to the toxic effects of palmitate treatment. The inhibition of lysosomal acid hydrolase activity by weak base treatment of islets under glucolipotoxic conditions causes islet degeneration that is prevented by the inhibition of protein palmitoylation. These findings indicate that defects in lysosomal function lead to the enhanced sensitivity of insulin-producing cells to palmitate and support a role for normal lysosomal function in the protection of ß cells from excess palmitate.


Asunto(s)
Cationes/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Manosafosfatos/metabolismo , Palmitatos/farmacología , Animales , Bovinos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Endocitosis/efectos de los fármacos , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/metabolismo , Células HEK293 , Humanos , Insulina/metabolismo , Lipoilación/efectos de los fármacos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Masculino , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
12.
Antioxid Redox Signal ; 29(14): 1432-1445, 2018 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-28978225

RESUMEN

SIGNIFICANCE: Cytokines released in and around pancreatic islets during islet inflammation are believed to contribute to impaired ß cell function and ß cell death during the development of diabetes. Nitric oxide, produced by ß cells in response to cytokine exposure, controls many of the responses of ß cells during islet inflammation. Recent Advances: Although nitric oxide has been shown to inhibit insulin secretion and oxidative metabolism and induce DNA damage in ß cells, it also activates protective pathways that promote recovery of insulin secretion and oxidative metabolism and repair of damaged DNA. Recent studies have identified a novel role for nitric oxide in selectively regulating the DNA damage response in ß cells. CRITICAL ISSUES: Does nitric oxide mediate cytokine-induced ß cell damage, or is nitric oxide produced by ß cells in response to cytokines to protect ß cells from damage? FUTURE DIRECTIONS: ß cells appear to be the only islet endocrine cell type capable of responding to proinflammatory cytokines with the production of nitric oxide, and these terminally differentiated cells have a limited capacity to regenerate. It is likely that there is a physiological purpose for this response, and understanding this could open new areas of study regarding the loss of functional ß cell mass during diabetes development.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Animales , Daño del ADN , Humanos , Óxido Nítrico/metabolismo
13.
Mol Cell Biol ; 36(15): 2067-77, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27185882

RESUMEN

Nitric oxide, produced in pancreatic ß cells in response to proinflammatory cytokines, plays a dual role in the regulation of ß-cell fate. While nitric oxide induces cellular damage and impairs ß-cell function, it also promotes ß-cell survival through activation of protective pathways that promote ß-cell recovery. In this study, we identify a novel mechanism in which nitric oxide prevents ß-cell apoptosis by attenuating the DNA damage response (DDR). Nitric oxide suppresses activation of the DDR (as measured by γH2AX formation and the phosphorylation of KAP1 and p53) in response to multiple genotoxic agents, including camptothecin, H2O2, and nitric oxide itself, despite the presence of DNA damage. While camptothecin and H2O2 both induce DDR activation, nitric oxide suppresses only camptothecin-induced apoptosis and not H2O2-induced necrosis. The ability of nitric oxide to suppress the DDR appears to be selective for pancreatic ß cells, as nitric oxide fails to inhibit DDR signaling in macrophages, hepatocytes, and fibroblasts, three additional cell types examined. While originally described as the damaging agent responsible for cytokine-induced ß-cell death, these studies identify a novel role for nitric oxide as a protective molecule that promotes ß-cell survival by suppressing DDR signaling and attenuating DNA damage-induced apoptosis.


Asunto(s)
Camptotecina/farmacología , Reparación del ADN/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Óxido Nítrico/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Supervivencia Celular , Daño del ADN/efectos de los fármacos , Células Hep G2 , Humanos , Células Secretoras de Insulina/citología , Masculino , Ratones , Especificidad de Órganos , Fosforilación/efectos de los fármacos , Células RAW 264.7 , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
14.
Am J Physiol Regul Integr Comp Physiol ; 309(5): R525-34, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26084699

RESUMEN

While insulinoma cells have been developed and proven to be extremely useful in studies focused on mechanisms controlling ß-cell function and viability, translating findings to human ß-cells has proven difficult because of the limited access to human islets and the absence of suitable insulinoma cell lines of human origin. Recently, a human ß-cell line, EndoC-ßH1, has been derived from human fetal pancreatic buds. The purpose of this study was to determine whether human EndoC-ßH1 cells respond to cytokines in a fashion comparable to human islets. Unlike most rodent-derived insulinoma cell lines that respond to cytokines in a manner consistent with rodent islets, EndoC-ßH1 cells fail to respond to a combination of cytokines (IL-1, IFN-γ, and TNF) in a manner consistent with human islets. Nitric oxide, produced following inducible nitric oxide synthase (iNOS) expression, is a major mediator of cytokine-induced human islet cell damage. We show that EndoC-ßH1 cells fail to express iNOS or produce nitric oxide in response to this combination of cytokines. Inhibitors of iNOS prevent cytokine-induced loss of human islet cell viability; however, they do not prevent cytokine-induced EndoC-ßH1 cell death. Stressed human islets or human islets expressing heat shock protein 70 (HSP70) are resistant to cytokines, and, much like stressed human islets, EndoC-ßH1 cells express HSP70 under basal conditions. Elevated basal expression of HSP70 in EndoC-ßH1 cells is consistent with the lack of iNOS expression in response to cytokine treatment. While expressing HSP70, EndoC-ßH1 cells fail to respond to endoplasmic reticulum stress activators, such as thapsigargin. These findings indicate that EndoC-ßH1 cells do not faithfully recapitulate the response of human islets to cytokines. Therefore, caution should be exercised when making conclusions regarding the actions of cytokines on human islets when using this human-derived insulinoma cell line.


Asunto(s)
Citocinas/farmacología , Mediadores de Inflamación/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Insulinoma/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Neoplasias Pancreáticas/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Ciclooxigenasa 2/metabolismo , Metabolismo Energético/efectos de los fármacos , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Insulinoma/patología , Interferón gamma/farmacología , Interleucina-1beta/farmacología , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Masculino , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Neoplasias Pancreáticas/patología , Fenotipo , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Factor de Necrosis Tumoral alfa/farmacología
15.
Stem Cells Transl Med ; 4(5): 483-93, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25834119

RESUMEN

The tumorigenic potential of human pluripotent stem cells (hPSCs) is a major limitation to the widespread use of hPSC derivatives in the clinic. Here, we demonstrate that the small molecule STF-31 is effective at eliminating undifferentiated hPSCs across a broad range of cell culture conditions with important advantages over previously described methods that target metabolic processes. Although STF-31 was originally described as an inhibitor of glucose transporter 1, these data support the reclassification of STF-31 as a specific NAD⁺ salvage pathway inhibitor through the inhibition of nicotinamide phosphoribosyltransferase (NAMPT). These findings demonstrate the importance of an NAD⁺ salvage pathway in hPSC biology and describe how inhibition of NAMPT can effectively eliminate hPSCs from culture. These results will advance and accelerate the development of safe, clinically relevant hPSC-derived cell-based therapies.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , NAD/antagonistas & inhibidores , Células Madre Pluripotentes/efectos de los fármacos , Piridinas/farmacología , Técnicas de Cultivo de Célula , Citocinas/antagonistas & inhibidores , Humanos , NAD/metabolismo , Nicotinamida Fosforribosiltransferasa/antagonistas & inhibidores , Células Madre Pluripotentes/citología , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología
16.
J Biol Chem ; 290(12): 7952-60, 2015 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-25648890

RESUMEN

Cytokines impair the function and decrease the viability of insulin-producing ß-cells by a pathway that requires the expression of inducible nitric oxide synthase (iNOS) and generation of high levels of nitric oxide. In addition to nitric oxide, excessive formation of reactive oxygen species, such as superoxide and hydrogen peroxide, has been shown to cause ß-cell damage. Although the reaction of nitric oxide with superoxide results in the formation of peroxynitrite, we have shown that ß-cells do not have the capacity to produce this powerful oxidant in response to cytokines. When ß-cells are forced to generate peroxynitrite using nitric oxide donors and superoxide-generating redox cycling agents, superoxide scavenges nitric oxide and prevents the inhibitory and destructive actions of nitric oxide on mitochondrial oxidative metabolism and ß-cell viability. In this study, we show that the ß-cell response to nitric oxide is regulated by the location of superoxide generation. Nitric oxide freely diffuses through cell membranes, and it reacts with superoxide produced within cells and in the extracellular space, generating peroxynitrite. However, only when it is produced within cells does superoxide attenuate nitric oxide-induced mitochondrial dysfunction, gene expression, and toxicity. These findings suggest that the location of radical generation and the site of radical reactions are key determinants in the functional response of ß-cells to reactive oxygen species and reactive nitrogen species. Although nitric oxide is freely diffusible, its biological function can be controlled by the local generation of superoxide, such that when this reaction occurs within ß-cells, superoxide protects ß-cells by scavenging nitric oxide.


Asunto(s)
Islotes Pancreáticos/metabolismo , Óxido Nítrico/metabolismo , Superóxidos/metabolismo , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN , Islotes Pancreáticos/citología , Ratones , Nitrógeno/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
17.
J Biol Chem ; 289(16): 11454-11464, 2014 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-24610783

RESUMEN

In this study, the effects of cytokines on the activation of the DNA double strand break repair factors histone H2AX (H2AX) and ataxia telangiectasia mutated (ATM) were examined in pancreatic ß cells. We show that cytokines stimulate H2AX phosphorylation (γH2AX formation) in rat islets and insulinoma cells in a nitric oxide- and ATM-dependent manner. In contrast to the well documented role of ATM in DNA repair, ATM does not appear to participate in the repair of nitric oxide-induced DNA damage. Instead, nitric oxide-induced γH2AX formation correlates temporally with the onset of irreversible DNA damage and the induction of apoptosis. Furthermore, inhibition of ATM attenuates cytokine-induced caspase activation. These findings show that the formation of DNA double strand breaks correlates with ATM activation, irreversible DNA damage, and ATM-dependent induction of apoptosis in cytokine-treated ß cells.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Células Secretoras de Insulina/metabolismo , Óxido Nítrico/metabolismo , Animales , Apoptosis/fisiología , Proteínas de la Ataxia Telangiectasia Mutada/genética , Caspasas/genética , Caspasas/metabolismo , Línea Celular Tumoral , Citocinas/genética , Citocinas/metabolismo , Roturas del ADN de Doble Cadena , Activación Enzimática/fisiología , Histonas , Células Secretoras de Insulina/citología , Masculino , Óxido Nítrico/genética , Fosfoproteínas , Fosforilación/fisiología , Ratas , Ratas Sprague-Dawley
18.
Vitam Horm ; 95: 299-322, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24559923

RESUMEN

Autoimmune diabetes is characterized by the selective destruction of insulin-secreting ß-cells that occurs during an inflammatory reaction in and around pancreatic islets of Langerhans. Cytokines such as interleukin-1, released by activated immune cells, have been shown to inhibit insulin secretion from pancreatic ß-cells and cause islet destruction. In response to cytokines, ß-cells express inducible nitric oxide synthase and produce micromolar levels of the free radical nitric oxide. Nitric oxide inhibits the mitochondrial oxidation of glucose resulting in an impairment of insulin secretion. Nitric oxide is also responsible for cytokine-mediated DNA damage in ß-cells. While nitric oxide mediates the inhibitory and toxic effects of cytokines, it also activates protective pathways that allow ß-cells to recover from this damage. This review will focus on the dual role of nitric oxide as a mediator of cytokine-induced damage and the activator of repair mechanisms that protect ß-cells from cytokine-mediated injury.


Asunto(s)
Estrés del Retículo Endoplásmico , Células Secretoras de Insulina/metabolismo , Modelos Biológicos , Óxido Nítrico/metabolismo , Animales , Apoptosis , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/patología , Necrosis , Óxido Nítrico Sintasa de Tipo II/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...