Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Cell ; 140(2): 222-34, 2010 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-20141836

RESUMEN

N-methyl-D-aspartate (NMDA) receptors constitute a major subtype of glutamate receptors at extrasynaptic sites that link multiple intracellular catabolic processes responsible for irreversible neuronal death. Here, we report that cerebral ischemia recruits death-associated protein kinase 1 (DAPK1) into the NMDA receptor NR2B protein complex in the cortex of adult mice. DAPK1 directly binds with the NMDA receptor NR2B C-terminal tail consisting of amino acid 1292-1304 (NR2B(CT)). A constitutively active DAPK1 phosphorylates NR2B subunit at Ser-1303 and in turn enhances the NR1/NR2B receptor channel conductance. Genetic deletion of DAPK1 or administration of NR2B(CT) that uncouples an activated DAPK1 from an NMDA receptor NR2B subunit in vivo in mice blocks injurious Ca(2+) influx through NMDA receptor channels at extrasynaptic sites and protects neurons against cerebral ischemic insults. Thus, DAPK1 physically and functionally interacts with the NMDA receptor NR2B subunit at extrasynaptic sites and this interaction acts as a central mediator for stroke damage.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Isquemia Encefálica/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Accidente Cerebrovascular/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Proteínas Reguladoras de la Apoptosis/genética , Encéfalo/metabolismo , Encéfalo/patología , Isquemia Encefálica/tratamiento farmacológico , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Muerte Celular , Proteínas Quinasas Asociadas a Muerte Celular , Ratones , Neuronas/citología , Neuronas/metabolismo , Péptidos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/patología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo
2.
J Pharmacol Exp Ther ; 376(3): 428-435, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33443084

RESUMEN

Nephrotic syndrome (NS) is associated with metabolic perturbances including profound dyslipidemia characterized by hypercholesterolemia and hypertriglyceridemia. A major underlying mechanism of hypertriglyceridemia in NS is lipoprotein lipase (LPL) deficiency and dysfunction. There is emerging evidence that elevated angiopoietin-like protein 3 (ANGPTL3), an LPL inhibitor that is primarily expressed and secreted by hepatocytes, may be in part responsible for these findings. Furthermore, there is evidence pointing to the contribution of ANGPTL3 to the pathogenesis of proteinuria in NS. Therefore, we hypothesized that inhibition of hepatic ANGPTL3 by RNA interference will ameliorate dyslipidemia and other symptoms of NS and pave the way for a new therapeutic strategy. To this end, we used a subcutaneously delivered, GalNAc (N-Acetylgalactosamine)-conjugated small interfering RNA (siRNA) to selectively target and suppress liver Angptl3 in rats with puromycin-induced NS, which exhibits clinical features of NS including proteinuria, hypoalbuminemia, hyperlipidemia, and renal histologic abnormalities. The study demonstrated that siRNA-mediated knockdown of the liver Angptl3 relieved its inhibitory effect on LPL and significantly reduced hypertriglyceridemia in nephrotic rats. This was accompanied by diminished proteinuria and hypoalbuminemia, which are the hallmarks of NS, and significant attenuation of renal tissue inflammation and oxidative stress. Taken together, this study confirmed the hypothesis that suppression of Angptl3 is protective in NS and points to the possibility that the use of RNA interference to suppress hepatic Angptl3 can serve as a novel therapeutic strategy for NS. SIGNIFICANCE STATEMENT: The current standard of care for mitigating nephrotic dyslipidemia in nephrotic syndrome is statins therapy. However, the efficacy of statins and its safety in the context of impaired kidney function is not well established. Here, we present an alternate therapeutic approach by using siRNA targeting Angptl3 expressed in hepatocytes. As the liver is the major source of circulating Angptl3, siRNA treatment reduced the profound hypertriglyceridemia in a rat model of nephrotic syndrome and was also effective in improving kidney and cardiac function.


Asunto(s)
Hipertrigliceridemia/complicaciones , Hígado/metabolismo , Síndrome Nefrótico/genética , Síndrome Nefrótico/prevención & control , Interferencia de ARN , Animales , Modelos Animales de Enfermedad , Lipoproteína Lipasa/metabolismo , Masculino , Síndrome Nefrótico/complicaciones , Ratas , Ratas Sprague-Dawley
4.
Circ Res ; 114(4): 626-36, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24366168

RESUMEN

RATIONALE: Increasing evidence has shown that proper control of mitochondrial dynamics (fusion and fission) is required for high-capacity ATP production in the heart. Transcriptional coactivators, peroxisome proliferator-activated receptor γ coactivator-1 (PGC-1) α and PGC-1ß, have been shown to regulate mitochondrial biogenesis in the heart at the time of birth. The function of PGC-1 coactivators in the heart after birth has been incompletely understood. OBJECTIVE: Our aim was to assess the role of PGC-1 coactivators during postnatal cardiac development and in adult hearts in mice. METHODS AND RESULTS: Conditional gene targeting was used in mice to explore the role of PGC-1 coactivators during postnatal cardiac development and in adult hearts. Marked mitochondrial structural derangements were observed in hearts of PGC-1α/ß-deficient mice during postnatal growth, including fragmentation and elongation, associated with the development of a lethal cardiomyopathy. The expression of genes involved in mitochondrial fusion (Mfn1, Opa1) and fission (Drp1, Fis1) was altered in the hearts of PGC-1α/ß-deficient mice. PGC-lα was shown to directly regulate Mfn1 gene transcription by coactivating the estrogen-related receptor α on a conserved DNA element. Surprisingly, PGC-1α/ß deficiency in the adult heart did not result in evidence of abnormal mitochondrial dynamics or heart failure. However, transcriptional profiling demonstrated that PGC-1 coactivators are required for high-level expression of nuclear- and mitochondrial-encoded genes involved in mitochondrial dynamics and energy transduction in the adult heart. CONCLUSIONS: These results reveal distinct developmental stage-specific programs involved in cardiac mitochondrial dynamics.


Asunto(s)
Cardiomiopatías/metabolismo , Corazón/crecimiento & desarrollo , Mitocondrias Cardíacas/metabolismo , Factores de Transcripción/metabolismo , Factores de Edad , Animales , Cardiomiopatías/genética , Progresión de la Enfermedad , Metabolismo Energético/fisiología , Receptor alfa de Estrógeno/metabolismo , Femenino , GTP Fosfohidrolasas/genética , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Ratones Noqueados , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Factores de Transcripción/genética
5.
Proc Natl Acad Sci U S A ; 109(8): E524-32, 2012 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-22315413

RESUMEN

Postprandial insulin release is regulated by glucose, but other circulating nutrients may target beta cells and potentiate glucose-stimulated insulin secretion via distinct signaling pathways. We demonstrate that fructose activates sweet taste receptors (TRs) on beta cells and synergizes with glucose to amplify insulin release in human and mouse islets. Genetic ablation of the sweet TR protein T1R2 obliterates fructose-induced insulin release and its potentiating effects on glucose-stimulated insulin secretion in vitro and in vivo. TR signaling in beta cells is triggered, at least in part, in parallel with the glucose metabolic pathway and leads to increases in intracellular calcium that are dependent on the activation of phospholipase C (PLC) and transient receptor potential cation channel, subfamily M, member 5 (TRPM5). Our results unveil a pathway for the regulation of insulin release by postprandial nutrients that involves beta cell sweet TR signaling.


Asunto(s)
Fructosa/farmacología , Glucosa/farmacología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Gusto/efectos de los fármacos , Animales , Calcio/metabolismo , Activación Enzimática/efectos de los fármacos , Eliminación de Gen , Humanos , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/enzimología , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Masculino , Ratones , Periodo Posprandial/efectos de los fármacos , Canales Catiónicos TRPM/metabolismo , Fosfolipasas de Tipo C/metabolismo
6.
Biomolecules ; 13(2)2023 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-36830621

RESUMEN

Several population studies have observed lower serum bilirubin levels in patients with non-alcoholic fatty liver disease (NAFLD). Yet, treatments to target this metabolic phenotype have not been explored. Therefore, we designed an N-Acetylgalactosamine (GalNAc) labeled RNAi to target the enzyme that clears bilirubin from the blood, the UGT1A1 glucuronyl enzyme (GNUR). In this study, male C57BL/6J mice were fed a high-fat diet (HFD, 60%) for 30 weeks to induce NAFLD and were treated subcutaneously with GNUR or sham (CTRL) once weekly for six weeks while continuing the HFD. The results show that GNUR treatments significantly raised plasma bilirubin levels and reduced plasma levels of the bilirubin catabolized product, urobilin. We show that GNUR decreased liver fat content and ceramide production via lipidomics and lowered fasting blood glucose and insulin levels. We performed extensive kinase activity analyses using our PamGene PamStation kinome technology and found a reorganization of the kinase pathways and a significant decrease in inflammatory mediators with GNUR versus CTRL treatments. These results demonstrate that GNUR increases plasma bilirubin and reduces plasma urobilin, reducing NAFLD and inflammation and improving overall liver health. These data indicate that UGT1A1 antagonism might serve as a treatment for NAFLD and may improve obesity-associated comorbidities.


Asunto(s)
Resistencia a la Insulina , Enfermedad del Hígado Graso no Alcohólico , Ratones , Animales , Masculino , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Urobilina/metabolismo , Bilirrubina , Ratones Endogámicos C57BL , Hígado/metabolismo , Transducción de Señal , Lípidos , Resistencia a la Insulina/genética
7.
Biomedicines ; 10(1)2022 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-35052799

RESUMEN

BACKGROUND: Saccharin is a common artificial sweetener and a bona fide ligand for sweet taste receptors (STR). STR can regulate insulin secretion in beta cells, so we investigated whether saccharin can stimulate insulin secretion dependent on STR and the activation of phospholipase C (PLC) signaling. METHODS: We performed in vivo and in vitro approaches in mice and cells with loss-of-function of STR signaling and specifically assessed the involvement of a PLC signaling cascade using real-time biosensors and calcium imaging. RESULTS: We found that the ingestion of a physiological amount of saccharin can potentiate insulin secretion dependent on STR. Similar to natural sweeteners, saccharin triggers the activation of the PLC signaling cascade, leading to calcium influx and the vesicular exocytosis of insulin. The effects of saccharin also partially require transient receptor potential cation channel M5 (TRPM5) activity. CONCLUSIONS: Saccharin ingestion may transiently potentiate insulin secretion through the activation of the canonical STR signaling pathway. These physiological effects provide a framework for understanding the potential health impact of saccharin use and the contribution of STR in peripheral tissues.

8.
Neuron ; 49(5): 719-33, 2006 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-16504947

RESUMEN

ADAR2 is a nuclear enzyme essential for GluR2 pre-mRNA editing at Q/R site-607, which gates Ca2+ entry through AMPA receptor channels. Here, we show that forebrain ischemia in adult rats selectively reduces expression of ADAR2 enzyme and, hence, disrupts RNA Q/R site editing of GluR2 subunit in vulnerable neurons. Recovery of GluR2 Q/R site editing by expression of exogenous ADAR2b gene or a constitutively active CREB, VP16-CREB, which induces expression of endogenous ADAR2, protects vulnerable neurons in the rat hippocampus from forebrain ischemic insult. Generation of a stable ADAR2 gene silencing by delivering small interfering RNA (siRNA) inhibits GluR2 Q/R site editing, leading to degeneration of ischemia-insensitive neurons. Direct introduction of the Q/R site edited GluR2 gene, GluR2(R607), rescues ADAR2 degeneration. Thus, ADAR2-dependent GluR2 Q/R site editing determines vulnerability of neurons in the rat hippocampus to forebrain ischemia.


Asunto(s)
Adenosina Desaminasa/metabolismo , Ataque Isquémico Transitorio/patología , Neuronas/citología , Prosencéfalo/citología , Edición de ARN/fisiología , Receptores AMPA/metabolismo , Animales , Animales Recién Nacidos , Northern Blotting/métodos , Western Blotting/métodos , Proteína de Unión a CREB/metabolismo , Calcio/metabolismo , Recuento de Células/métodos , Supervivencia Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Estimulación Eléctrica/métodos , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/citología , Inmunohistoquímica/métodos , Técnicas In Vitro , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Potenciales de la Membrana/efectos de la radiación , Neuronas/metabolismo , Fosfopiruvato Hidratasa/metabolismo , ARN Mensajero/biosíntesis , ARN Interferente Pequeño/farmacología , Proteínas de Unión al ARN , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos
9.
J Biol Chem ; 284(38): 26070-84, 2009 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-19628872

RESUMEN

Poly(ADP-ribose) polymerase-1 (PARP-1) has been implicated in the pathogenesis of several central nervous system (CNS) disorders. However, the role of PARP-1 in autoimmune CNS injury remains poorly understood. Therefore, we studied experimental autoimmune encephalomyelitis (EAE), a model for multiple sclerosis in mice with a targeted deletion of PARP-1. We identified inherent physiological abnormalities in the circulating and splenic immune composition between PARP-1(-/-) and wild type (WT) mice. Upon EAE induction, PARP-1(-/-) mice had an earlier onset and developed a more severe EAE compared with WT cohorts. Splenic response was significantly higher in PARP-1(-/-) mice largely because of B cell expansion. Although formation of Th1 and Th17 effector T lymphocytes was unaffected, PARP-1(-/-) mice had significantly earlier CD4+ T lymphocyte and macrophage infiltration into the CNS during EAE. However, we did not detect significant differences in cytokine profiles between PARP-1(-/-) and WT spinal cords at the peak of EAE. Expression analysis of different PARP isozymes in EAE spinal cords showed that PARP-1 was down-regulated in WT mice and that PARP-3 but not PARP-2 was dramatically up-regulated in both PARP-1(-/-) and WT mice, suggesting that these PARP isozymes could have distinct roles in different CNS pathologies. Together, our results indicate that PARP-1 plays an important role in regulating the physiological immune composition and in immune modulation during EAE; our finding identifies a new aspect of immune regulation by PARPs in autoimmune CNS pathology.


Asunto(s)
Encefalomielitis Autoinmune Experimental/enzimología , Esclerosis Múltiple/enzimología , Poli(ADP-Ribosa) Polimerasas , Médula Espinal/enzimología , Animales , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Regulación Enzimológica de la Expresión Génica/genética , Regulación Enzimológica de la Expresión Génica/inmunología , Isoenzimas/biosíntesis , Isoenzimas/genética , Isoenzimas/inmunología , Macrófagos/enzimología , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Noqueados , Esclerosis Múltiple/genética , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/biosíntesis , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/inmunología , Médula Espinal/inmunología , Médula Espinal/patología , Células TH1/enzimología , Células TH1/inmunología , Células TH1/patología , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
10.
PLoS One ; 15(7): e0236892, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32735622

RESUMEN

There is a significant unmet need for a safe and effective therapy for the treatment of children with congenital hyperinsulinism. We hypothesized that amplification of the glucagon signaling pathway could ameliorate hyperinsulinism associated hypoglycemia. In order to test this we evaluated the effects of loss of Prkar1a, a negative regulator of Protein Kinase A in the context of hyperinsulinemic conditions. With reduction of Prkar1a expression, we observed a significant upregulation of hepatic gluconeogenic genes. In wild type mice receiving a continuous infusion of insulin by mini-osmotic pump, we observed a 2-fold increase in the level of circulating ketones and a more than 40-fold increase in Kiss1 expression with reduction of Prkar1a. Loss of Prkar1a in the Sur1-/- mouse model of KATP hyperinsulinism significantly attenuated fasting induced hypoglycemia, decreased the insulin/glucose ratio, and also increased the hepatic expression of Kiss1 by more than 10-fold. Together these data demonstrate that amplification of the hepatic glucagon signaling pathway is able to rescue hypoglycemia caused by hyperinsulinism.


Asunto(s)
Hiperinsulinismo Congénito/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Kisspeptinas/genética , Receptores de Sulfonilureas/genética , Animales , Hiperinsulinismo Congénito/genética , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Gluconeogénesis/genética , Glucosa/metabolismo , Hipoglucemia/metabolismo , Insulina/metabolismo , Cetonas/metabolismo , Kisspeptinas/metabolismo , Hígado/metabolismo , Ratones , Ratones Noqueados , Transducción de Señal
11.
J Neurochem ; 103(5): 1721-9, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17944875

RESUMEN

ATP-sensitive K(+) (K(ATP)) channels that are gated by intracellular ATP/ADP concentrations are a unique subtype of potassium channels and play an essential role in coupling intracellular metabolic events to electrical activity. Opening of K(ATP) channels during energy deficits in the CNS induces efflux of potassium ions and in turn hyperpolarizes neurons. Thus, activation of K(ATP) channels is thought to be able to counteract excitatory insults and protect against neuronal death. In this review, we bring together recent studies about what kinds of molecules are needed to build and regulate arrays of K(ATP) channel functions in the CNS neurons. We propose a model to explain how K(ATP) channel activation regulates glutamate release from the pre-synaptic terminals and how this regulation protects against ischemic neuronal injury and epilepsy.


Asunto(s)
Degeneración Nerviosa/prevención & control , Neuronas/fisiología , Canales de Potasio de Rectificación Interna/metabolismo , Animales , Ácido Glutámico/metabolismo , Ácido Glutámico/toxicidad , Activación del Canal Iónico , Modelos Biológicos , Degeneración Nerviosa/metabolismo , Neuronas/efectos de los fármacos , Canales de Potasio de Rectificación Interna/genética , Receptores de N-Metil-D-Aspartato/fisiología , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
12.
J Neurochem ; 103(5): 1982-8, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17883401

RESUMEN

The Kir6.1 channels are a subtype of ATP-sensitive inwardly rectifying potassium (K(ATP)) channels that play an essential role in coupling the cell's metabolic events to electrical activity. In this study, we show that functional Kir6.1 channels are located at excitatory pre-synaptic terminals as a complex with type-1 Sulfonylurea receptors (SUR1) in the hippocampus. The mutant mice with deficiencies in expressing the Kir6.1 or the SUR1 gene are more vulnerable to generation of epileptic form of seizures, compared to wild-type controls. Whole-cell patch clamp recordings demonstrate that genetic deletion of the Kir6.1/SUR1 channels enhances glutamate release at CA3 synapses. Hence, expression of functional Kir6.1/SUR1 channels inhibits seizure responses and possibly acts via limiting excitatory glutamate release.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Ácido Glutámico/metabolismo , Hipocampo/citología , Canales de Potasio de Rectificación Interna/fisiología , Convulsiones/etiología , Sinapsis/metabolismo , Animales , Conducta Animal , Agonistas de Aminoácidos Excitadores/farmacología , Hipocampo/metabolismo , Humanos , Inmunoprecipitación/métodos , Técnicas In Vitro , Canales KATP , Ácido Kaínico/farmacología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/efectos de la radiación , Ratones , Ratones Noqueados , Técnicas de Placa-Clamp/métodos , Canales de Potasio de Rectificación Interna/deficiencia , Ratas , Ratas Sprague-Dawley , Convulsiones/genética , Convulsiones/fisiopatología , Sinapsis/efectos de los fármacos
13.
PLoS One ; 12(8): e0182371, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28796801

RESUMEN

Pregnancy is associated with increased ß-cell proliferation driven by prolactin. Long noncoding RNAs (lncRNA) are the most abundant RNA species in the mammalian genome, yet, their functional importance is mainly elusive. AIMS/HYPOTHESIS: This study tests the hypothesis that lncRNAs regulate ß-cell proliferation in response to prolactin in the context of ß-cell mass compensation in pregnancy. METHODS: The expression profile of lncRNAs in mouse islets at day 14.5 of pregnancy was explored by a bioinformatics approach, further confirmed by quantitative PCR at different days of pregnancy, and islet specificity was evaluated by comparing expression in islets versus other tissues. In order to establish the role of the candidate lncRNAs we studied cell proliferation in mouse islets and the MIN6 ß-cell line by EdU incorporation and cell count. RESULTS: We found that a group of lncRNAs is differentially regulated in mouse islets at 14.5 days of pregnancy. At different stages of pregnancy, these lncRNAs are dynamically expressed, and expression is prolactin dependent in mouse islets and MIN6 cells. One of those lncRNAs, Gm16308 (Lnc03), is dynamically regulated during pregnancy, prolactin-dependent and islet-enriched. Silencing Lnc03 in primary ß-cells and MIN6 cells inhibits, whereas over-expression stimulates, proliferation even in the absence of prolactin, demonstrating that Lnc03 regulates ß-cell growth. CONCLUSIONS/INTERPRETATION: During pregnancy mouse islet proliferation is correlated with dynamic changes of lncRNA expression. In particular, Lnc03 regulates mouse ß-cell proliferation and may be a crucial component of ß-cell proliferation in ß-cell mass adaptation in both health and disease.


Asunto(s)
Proliferación Celular , Células Secretoras de Insulina/fisiología , ARN Largo no Codificante/fisiología , Animales , Células Cultivadas , Femenino , Regulación del Desarrollo de la Expresión Génica , Ratones Endogámicos C57BL , Embarazo , Prolactina/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Transcriptoma
14.
J Clin Invest ; 126(9): 3567-79, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27500491

RESUMEN

Intramuscular lipid accumulation is a common manifestation of chronic caloric excess and obesity that is strongly associated with insulin resistance. The mechanistic links between lipid accumulation in myocytes and insulin resistance are not completely understood. In this work, we used a high-throughput chemical biology screen to identify a small-molecule probe, SBI-477, that coordinately inhibited triacylglyceride (TAG) synthesis and enhanced basal glucose uptake in human skeletal myocytes. We then determined that SBI-477 stimulated insulin signaling by deactivating the transcription factor MondoA, leading to reduced expression of the insulin pathway suppressors thioredoxin-interacting protein (TXNIP) and arrestin domain-containing 4 (ARRDC4). Depleting MondoA in myocytes reproduced the effects of SBI-477 on glucose uptake and myocyte lipid accumulation. Furthermore, an analog of SBI-477 suppressed TXNIP expression, reduced muscle and liver TAG levels, enhanced insulin signaling, and improved glucose tolerance in mice fed a high-fat diet. These results identify a key role for MondoA-directed programs in the coordinated control of myocyte lipid balance and insulin signaling and suggest that this pathway may have potential as a therapeutic target for insulin resistance and lipotoxicity.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/fisiología , Insulina/metabolismo , Metabolismo de los Lípidos , Células Musculares/metabolismo , Músculo Esquelético/citología , Animales , Arrestinas/metabolismo , Biopsia , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Dieta Alta en Grasa , Glucosa/metabolismo , Homeostasis , Resistencia a la Insulina/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas , Transducción de Señal , Tiorredoxinas/metabolismo , Transfección , Triglicéridos/metabolismo
15.
Mol Neurobiol ; 32(2): 145-55, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16215279

RESUMEN

Ischemic stroke, or a brain attack, is the third leading cause of death in developed countries. A critical feature of the disease is a highly selective pattern of neuronal loss; certain identifiable subsets of neurons--particularly CA1 pyramidal neurons in the hippocampus are severely damaged, whereas others remain intact. A key step in this selective neuronal injury is Ca2+/Zn2+ entry into vulnerable neurons through alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor channels, a principle subtype of glutamate receptors. AMPA receptor channels are assembled from glutamate receptor (GluR)1, -2, -3, and -4 subunits. Circumstance data have indicated that the GluR2 subunits dictate Ca2+/Zn2+ permeability of AMPA receptor channels and gate injurious Ca2+/Zn2+ signals in vulnerable neurons. Therefore, targeting to the AMPA receptor subunit GluR2 can be considered a practical strategy for stroke therapy.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Receptores AMPA/metabolismo , Glutamatos/toxicidad , Humanos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , Receptores AMPA/genética , Receptores de N-Metil-D-Aspartato/metabolismo
16.
J Biomol Screen ; 19(1): 77-87, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23989452

RESUMEN

Excess caloric consumption leads to triacylglyceride (TAG) accumulation in tissues that do not typically store fat, such as skeletal muscle. This ectopic accumulation alters cells, contributing to the pathogenesis of metabolic syndrome, a major health problem worldwide. We developed a 1536-well assay to measure intracellular TAG accumulation in differentiating H9c2 myoblasts. For this assay, cells were incubated with oleic acid to stimulate TAG accumulation prior to adding compounds. We used Nile red as a fluorescent dye to quantify TAG content with a microplate reader. The cell nuclei were counterstained with DAPI nuclear stain to assess cell count and filter cytotoxic compounds. In parallel, we developed an image-based assay in H9c2 cells to measure lipid accumulation levels via high-content analysis, exploiting the dual-emission spectra characteristic of Nile red staining of neutral and phospholipids. Using both approaches, we successfully screened ~227,000 compounds from the National Institutes of Health library. The screening data from the plate reader and IC50 values correlated with that from the Opera QEHS cell imager. The 1536-well plate reader assay is a powerful high-throughout screening platform to identify potent inhibitors of TAG accumulation to better understand the molecular pathways involved in lipid metabolism that lead to lipotoxicity.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Ensayos Analíticos de Alto Rendimiento , Metabolismo de los Lípidos/efectos de los fármacos , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Triglicéridos/metabolismo , Técnicas de Cultivo de Célula , Línea Celular , Descubrimiento de Drogas/métodos , Humanos , Reproducibilidad de los Resultados
17.
Sci Rep ; 1: 2, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22355521

RESUMEN

Basic helix-loop-helix transcription factors Olig1 and Olig2 critically regulate oligodendrocyte development. Initially identified as a downstream effector of Olig1, an oligodendrocyte-specific zinc finger transcription repressor, Zfp488, cooperates with Olig2 function. Although Zfp488 is required for oligodendrocyte precursor formation and differentiation during embryonic development, its role in oligodendrogenesis of adult neural progenitor cells is not known. In this study, we tested whether Zfp488 could promote an oligodendrogenic fate in adult subventricular zone (SVZ) neural stem/progenitor cells (NSPCs). Using a cuprizone-induced demyelination model in mice, we examined the effect of retrovirus-mediated Zfp488 overexpression in SVZ NSPCs. Our results showed that Zfp488 efficiently promoted the differentiation of the SVZ NSPCs into mature oligodendrocytes in vivo. After cuprizone-induced demyelination injury, Zfp488-transduced mice also showed significant restoration of motor function to levels comparable to control mice. Together, these findings identify a previously unreported role for Zfp488 in adult oligodendrogenesis and functional remyelination after injury.


Asunto(s)
Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/patología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Oligodendroglía/metabolismo , Oligodendroglía/patología , Proteínas Represoras/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Ratones , Ratones Noqueados
18.
Am J Physiol Renal Physiol ; 288(2): F371-9, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15454391

RESUMEN

Omi/HtrA2 is a mitochondrial proapoptotic serine protease that is able to induce both caspase-dependent and caspase-independent cell death. After apoptotic stimuli, Omi is released to the cytoplasm where it binds and cleaves inhibitor of apoptosis proteins. In this report, we investigated the role of Omi in renal cell death following cisplatin treatment. Using primary mouse proximal tubule cells, as well as established renal cell lines, we show that the level of Omi protein is upregulated after treatment with cisplatin. This upregulation is followed by the release of Omi from mitochondria to the cytoplasm and degradation of XIAP. Reducing the endogenous level of Omi protein using RNA interference renders renal cells resistant to cisplatin-induced cell death. Furthermore, we show that the proteolytic activity of Omi is necessary and essential for cisplatin-induced cell death in this system. When renal cells are treated with Omi's specific inhibitor, ucf-101, they become significantly resistant to cisplatin-induced cell death. Ucf-101 was also able to minimize cisplatin-induced nephrotoxic injury in animals. Our results demonstrate that Omi is a major mediator of cisplatin-induced cell death in renal cells and suggest a way to limit renal injury by specifically inhibiting its proteolytic activity.


Asunto(s)
Antineoplásicos/toxicidad , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Cisplatino/toxicidad , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , Serina Endopeptidasas/farmacología , Animales , Técnicas de Cultivo de Célula , Serina Peptidasa A2 que Requiere Temperaturas Altas , Humanos , Túbulos Renales Proximales/citología , Ratones , Ratones Endogámicos C57BL , Proteínas Mitocondriales , Proteínas/metabolismo
19.
J Biol Chem ; 279(48): 50295-301, 2004 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-15371414

RESUMEN

Omi/HtrA2 is a nuclear-encoded mitochondrial serine protease that has a pro-apoptotic function in mammalian cells. Upon induction of apoptosis, Omi translocates to the cytoplasm and participates in caspase-dependent apoptosis by binding and degrading inhibitor of apoptosis proteins. Omi can also initiate caspase-independent apoptosis in a process that relies entirely on its ability to function as an active protease. To investigate the mechanism of Omi-induced apoptosis, we set out to isolate novel substrates that are cleaved by this protease. We identified HS1-associated protein X-1 (HAX-1), a mitochondrial anti-apoptotic protein, as a specific Omi interactor that is cleaved by Omi both in vitro and in vivo. HAX-1 degradation follows Omi activation in cells treated with various apoptotic stimuli. Using a specific inhibitor of Omi, HAX-1 degradation is prevented and cell death is reduced. Cleavage of HAX-1 was not observed in a cell line derived from motor neuron degeneration 2 mice that carry a mutated form of Omi that affects its proteolytic activity. Degradation of HAX-1 is an early event in the apoptotic process and occurs while Omi is still confined in the mitochondria. Our results suggest that Omi has a unique pro-apoptotic function in mitochondria that involves removal of the HAX-1 anti-apoptotic protein. This function is distinct from its ability to activate caspase-dependent apoptosis in the cytoplasm by degrading inhibitor of apoptosis proteins.


Asunto(s)
Apoptosis/fisiología , Proteínas/metabolismo , Serina Endopeptidasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Serina Peptidasa A2 que Requiere Temperaturas Altas , Humanos , Péptidos y Proteínas de Señalización Intracelular , Potenciales de la Membrana/fisiología , Ratones , Mitocondrias/metabolismo , Proteínas Mitocondriales , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA